Entry - *305900 - GLUCOSE-6-PHOSPHATE DEHYDROGENASE; G6PD - OMIM
 
* 305900

GLUCOSE-6-PHOSPHATE DEHYDROGENASE; G6PD


HGNC Approved Gene Symbol: G6PD

Cytogenetic location: Xq28     Genomic coordinates (GRCh38): X:154,531,390-154,547,569 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
Xq28 {Resistance to malaria due to G6PD deficiency} 611162 3
Hemolytic anemia, G6PD deficient (favism) 300908 XL 3

TEXT

Description

Glucose-6-phosphate dehydrogenase (G6PD; EC 1.1.1.49) plays a key role in the production of ribose 5-phosphate and the generation of NADPH in the hexose monophosphate pathway. Because this pathway is the only NADPH-generation process in mature red cells, which lack the citric acid cycle, a genetic deficiency of G6PD (300908) is often associated with adverse physiologic effects (summary by Takizawa et al., 1986).


Cloning and Expression

Takizawa et al. (1986) cloned G6PD from a human hepatoma cDNA library. The deduced 531-amino acid protein has a molecular mass of 58 kD. Cappellini and Fiorelli (2008) stated that the G6PD protein contains 515 amino acids.


Gene Structure

Martini et al. (1986) determined that the human G6PD gene has 13 exons and spans 18 kb. The protein-coding region is divided into 12 segments, ranging from 12 to 236 bp, and an intron is present in the 5-prime untranslated region. The major 5-prime end of mature G6PD mRNA in several cell lines is located 177 bp upstream of the translation-initiating codon. Comparison of the promoter region of G6PD and 10 other housekeeping enzyme genes confirmed the presence of common features. In particular, in 8 cases in which a 'TATA' box was present, a conserved sequence of 25 bp was seen immediately downstream.

Chen et al. (1991) determined the sequence of 20,114 bp of human DNA including the G6PD gene. The region included a prominent CpG island, starting about 680 nucleotides upstream of the transcription initiation site, extending about 1,050 nucleotides downstream of the initiation site, and ending at the start of the first intron. The transcribed region from the initiation site to the poly(A) addition site covered 15,860 bp. The sequence of the 13 exons agreed with the published cDNA sequence and, for the 11 exons tested, with the corresponding sequence in a yeast artificial chromosome (YAC). Sixteen Alu sequences constituted 24% of the total sequence tract. Four were outside the borders of the mRNA transcript of the gene; all of the others were found in a large (9,858 bp) intron between exons 2 and 3.

The Japanese pufferfish Fugu rubripes is a useful model for the comparative study of vertebrate genomes because of the compact nature of its genome. Since the Fugu genome is approximately 8 times smaller than that of mammals, most genes should be more compact. To test this hypothesis, Mason et al. (1995) cloned and sequenced the G6PD gene from Fugu and compared it to the corresponding human gene. The intron/exon structure of the 2 genes was identical throughout the protein coding regions. Intron 2 is also the largest intron in both species. However, they found that the Fugu gene was 4 times smaller than the human gene; the difference was accounted for by the fact that the pufferfish gene has smaller introns. Mason et al. (1995) constructed a molecular phylogeny for 10 G6PD protein sequences. The sequences fell in the expected arrangement based on established phylogenetic relationships, with the Plasmodium falciparum sequence diverging most widely.

Fusco et al. (2012) stated that the G6PD gene, which is transcribed in the telomeric direction, partly overlaps the IKBKG gene (300248), which is transcribed in the centromeric direction. The genes share a conserved promoter region that has bidirectional housekeeping activity. In addition, intron 2 of the G6PD gene contains an alternate promoter for the IKBKG gene only. Fusco et al. (2012) determined that the region containing the G6PD gene and the 5-prime end of the IKBKG gene contains Alu elements.


Evolution

Notaro et al. (2000) showed that an evolutionary analysis is a key to understanding the biology of a housekeeping gene such as G6PD. From the alignment of the amino acid sequence of 52 G6PD species from 42 different organisms, they found a striking correlation between the amino acid replacements that cause G6PD deficiency in humans and the sequence conservation of G6PD. Two-thirds of such replacements were found in highly and moderately conserved (50 to 99%) amino acids; relatively few were located in fully conserved amino acids (where they might be lethal) or in poorly conserved amino acids (where presumably they simply would not cause G6PD deficiency). The findings were considered consistent with the notion that all human mutants have residual enzyme activity and that null mutations are lethal at some stage of development. Comparing the distribution of mutations in the human housekeeping gene with evolutionary conservation is a useful tool for pinpointing amino acid residues important for the stability or the function of the corresponding protein.


Mapping

Childs et al. (1958) determined that the G6PD gene resides on the X chromosome.

From study of radiation-induced segregants (irradiated human cells 'rescued' by fusion with hamster cells), Goss and Harris (1977) showed that the order of 4 loci on the X chromosome is PGK: alpha-GAL: HPRT: G6PD and that the 3 intervals between these 4 loci are, in relative terms, 0.33, 0.30, and 0.23.

Studying X-autosome translocations in somatic cell hybrids, Pai et al. (1980) showed that a breakpoint at the junction of Xq27-q28 separates HPRT from G6PD. G6PD is distally situated at Xq28. They localized HPRT to the segment between Xq26 and Xq27.

That G6PD is X-linked in the mouse was supported by Epstein's finding (1969) that oocytes of XO females have half as much G6PD as do oocytes of XX female mice. The level of lactate dehydrogenase was the same. Epstein's conclusion was that the G6PD gene is X-linked in the mouse, that synthesis occurs in the oocyte and is dosage-dependent, and that X inactivation does not occur in oocytes.


Gene Function

Ninfali et al. (1995) studied muscle expression of G6PD in normal individuals and in persons with G6PD deficiency of 3 types. They were prompted to undertake these studies because of patients with symptoms such as myalgia, cramps, and muscle weakness under conditions of stress, particularly physical exertion. All 3 variants--Mediterranean (305900.0006), Seattle-like (305900.0010), and G6PD A- (305900.0002)--showed the enzyme defect in muscle. A statistically significant relationship was found in the activity of G6PD in erythrocytes and muscle of male subjects. The results suggested to the authors that, for a given variant, the extent of the enzyme defect in muscle can be determined from the G6PD activity of erythrocytes, using an equation that they derived.

In studies in bovine aortic and human coronary artery endothelial cells, Leopold et al. (2007) demonstrated that aldosterone decreased G6PD expression and activity, resulting in increased oxidant stress and decreased nitric oxide levels, similar to what is observed in G6PD-deficient endothelial cells. Aldosterone decreased G6PD expression by increasing expression of the cAMP-response element modulator (CREM; 123812), thereby inhibiting cAMP-response element binding protein (CREB; 123810)-mediated G6PD transcription. In vivo aldosterone infusion in mice decreased vascular G6PD expression and impaired vascular reactivity; these effects were abrogated by spironolactone or vascular gene transfer of G6pd. Leopold et al. (2007) concluded that aldosterone induces a G6PD-deficient phenotype to impair endothelial function.


Population Genetics

Different variants of G6PD are found in high frequency in African, Mediterranean, and Asiatic populations (Porter et al., 1964), and heterozygote advantage vis-a-vis malaria (Luzzatto et al., 1969) has been invoked to account for the high frequency of the particular alleles in particular populations.


Molecular Genetics

The variety of forms of the G6PD enzyme is great (Yoshida et al., 1971; Beutler and Yoshida, 1973; Yoshida and Beutler, 1978). The World Health Organization (WHO (1967, 1967)) gave its attention to problems of nomenclature and standard procedures for study. The demonstrated polymorphism at this X-linked locus rivals that of the autosomal loci for the polypeptide chains of hemoglobin. As in the latter instance, single amino acid substitution has been demonstrated as the basis of the change in the G6PD molecule resulting from mutation (Yoshida et al., 1967).

The G6PD variants have been divided into 5 classes according to the level of enzyme activity: class 1--enzyme deficiency with chronic nonspherocytic hemolytic anemia; class 2--severe enzyme deficiency (less than 10%); class 3--moderate to mild enzyme deficiency (10-60%); class 4--very mild or no enzyme deficiency (60%); class 5--increased enzyme activity. Mutations causing nonspherocytic hemolytic anemia are clustered near the carboxy end of the enzyme, in the region between amino acids 362 and 446, whereas most of the clinically mild mutations are located at the amino end of the molecule. As the intragenic defects have been identified, many variants that were thought to be unique have been found to be identical on sequence analysis. This finding should not be surprising inasmuch as the methods of biochemical characterization are not very accurate, particularly when dealing with mutant enzymes that are unstable. For example, although the patients were unrelated, G6PD Marion, G6PD Gastonia, and G6PD Minnesota had the same val213-to-leu substitution; and G6PD Nashville and G6PD Anaheim were found to have the same arg393-to-his substitution (Beutler et al., 1991).

The frequencies of low-activity alleles of G6PD in humans are highly correlated with the prevalence of malaria (see 611162). These deficiency alleles are thought to provide reduced risk for infection by the Plasmodium parasite and are maintained at high frequency despite the illnesses that they cause. Haplotype analysis of A- (305900.0002) and Mediterranean (Med) (305900.0006) mutations at this locus indicates that they had evolved independently and have increased in frequency at a rate that is too rapid to be explained by random genetic drift. Tishkoff et al. (2001) used statistical modeling to demonstrate that the A- allele arose within a past 3840 to 11,760 years and the Med allele arose within the past 1600 to 6640 years. Tishkoff et al. (2001) concluded that their results support the hypothesis that malaria has had a major impact on humans only since the introduction of agriculture within the past 10,000 years and provide a striking example of the signature of selection on the human genome.

That resistance to severe malaria is the basis of the high frequency of G6PD deficiency and that both hemizygotes and heterozygotes enjoy an advantage was established by Ruwende et al. (1995) in 2 large case-control studies of more than 2,000 African children. They found that the common African form of G6PD deficiency (G6PD A-; 305900.0002) was associated with a 46 to 58% reduction in risk of severe malaria for both female heterozygotes and male hemizygotes. A mathematical model incorporating the measured selective advantage against malaria suggested that a counterbalancing selective disadvantage, associated with this enzyme deficiency, has retarded its rise in frequency in malaria-endemic regions.

Sansone et al. (1981) described 6 G6PD variants in Italian males, all associated with enzyme deficiency and 2 with signs of hemolysis. They provided a useful map of 19 sporadic G6PD variants found in Italy. They mapped to regions where the common forms of G6PD deficiency are frequent.

Hitzeroth and Bender (1981) found an increasing frequency of apparent BB homozygotes with increasing age of groups of South African blacks studied. They suggested that this represents selection against A- cell lines in heterozygotes and speculated further that malaria is the underlying selective agent.

Mohrenweiser and Neel (1981) identified thermolabile variants of lactate dehydrogenase B, glucosephosphate isomerase, and glucose-6-phosphate dehydrogenase. None was detectable as a variant by standard electrophoretic techniques. All were inherited. Beutler (1983) hypothesized that the marked differences in the extent to which various tissues manifest the deficiency state in various enzymopathies including G6PD deficiency may be related to tissue-to-tissue differences in proteases. Mutation may produce changes in susceptibility of the enzyme to proteases.

Data on gene frequencies of allelic variants were tabulated by Roychoudhury and Nei (1988).

Vulliamy et al. (1988) cloned and sequenced 7 mutant G6PD alleles. A single point mutation in the African variant G6PD A (305900.0001) does not result in deficiency of the enzyme. The other 6 mutants were all associated with enzyme deficiency. Single point mutations were identified in G6PD Mediterranean (305900.0006), G6PD Metaponto (305900.0007), G6PD Ilesha (305900.0004), G6PD Chatham (305900.0003), G6PD Santiago de Cuba (305900.0009), and G6PD Matera (an example of A-; 305900.0002).

By use of 14 unique sequence probes and 18 restriction enzymes, D'Urso et al. (1988) found a polymorphic silent mutation in the G6PD gene. A PstI site that maps to exon 10 was monomorphic in all British and Italian subjects studied, but was polymorphic in West African people. Specifically, it was absent from 22% of Nigerian X chromosomes. By sequence analysis, D'Urso et al. (1988) showed that the absence of this PstI site resulted from a G-to-A replacement at position 1116, corresponding to the third base of a glutamine codon (see 305900.0017). No amino acid change was produced in the protein. Yoshida et al. (1988) reported 2 RFLPs of the G6PD locus with high frequency in blacks and showed statistically significant linkage disequilibrium between the A+/B+ types and 1 of the RFLPs at the G6PD locus.

Vulliamy et al. (1988) found a striking predominance of C-to-T transitions among the G6PD mutations, with GC doublets involved in 4 of the 7 cases. It has been found that even in the same population, more than 1 G6PD variant is present. For example, in the island of Sardinia, extensive clinical and biochemical studies identified 4 different G6PD variants. De Vita et al. (1989) cloned and sequenced the 4 G6PD variants and found that at the molecular level there were only 2 mutations. The first mutation had an asp282-to-his change resulting from a GAT-to-CAT change in exon 8. This mutation caused the G6PD Seattle-like phenotype, a relatively mild form of G6PD deficiency (see 305900.0010). The other 3 variants were accompanied by very severe G6PD deficiency. All 3 had a ser188-to-phe change resulting from a TCC-to-TTC transition. This is the same change as that in G6PD Mediterranean (305900.0006). These 3 Sardinian variants also had a silent mutation in exon 11 with a change of TAC-to-TAT, both of which encode tyrosine at amino acid 437. These findings indicate that some G6PD-deficient variants identified only on the basis of their biochemical characteristics may not correspond to different mutations in the G6PD gene. The variations may be due to posttranscriptional or posttranslational modifications of the enzyme; whether the modifications are due to mutations in a tightly linked gene or to noninherited physiologic changes could not be distinguished with the data available. Study of families in which different forms of G6PD Mediterranean segregate suggested that the biochemical characteristics are transmitted in the family along with the enzyme deficiency, thus favoring the first hypothesis.

In a study of an unselected sample of 1,524 schoolboys from the province of Matera (Lucania) in southern Italy, Calabro et al. (1990) found that although the most frequent form of G6PD deficiency was G6PD Mediterranean, an extraordinary number of other forms existed. The overall rate of G6PD deficiency was 2.6%. The frequency ranged from 7.2% on the Ionian coast to zero on the eastern side of the Lucanian Apennines.

Kay et al. (1992) analyzed the evolution of the G6PD gene by examining the DNA samples from 54 male African Americans for G6PD A+ (305900.0001), G6PD A- (305900.0002), and G6PD B and for polymorphisms in intron 5 (PvuII), at nucleotide 1311 (305900.0018), and at nucleotide 1116 (305900.0017). They concluded from these and their previous studies that G6PD B is the most ancient genotype. The nucleotide 1311 mutation, with its worldwide distribution, probably occurred next. The PstI mutation, limited to Africans, probably arose next and is more ancient than the A+ mutation, which occurred in a gene without either the PstI or the 1311 mutation. G6PD A- (202A/376G) is the most recent mutation and is still in linkage disequilibrium with all of the sites. It presumably occurred in an individual with both the A+ and PvuII mutations.

Chiu et al. (1993) reported molecular characterization of the defects in 43 G6PD-deficient Chinese males whose G6PD had been well characterized biochemically. Among the 43 samples, they identified 5 different nucleotide substitutions: 1388G-A (arg to his; 305900.0029); 1376G-T (arg to leu; 305900.0021); 1024C-T (leu to phe; 305900.0046); 392G-T (gly to val; 305900.0045); and 95A-G (his to arg; 305900.0044). The 5 substitutions accounted for 36 of the 43 samples; none of these substitutions had been reported in non-Asians. The substitutions at nucleotides 392 and 1024 were new findings. The substitutions at nucleotides 1376 and 1388 accounted for over one-half of the samples.

Vulliamy et al. (1993) tabulated 58 different mutations in the G6PD gene that account for 97 named G6PD variants. The mutations were almost exclusively missense mutations, causing single amino acid substitutions. They were spread throughout the coding region of the gene, although there appeared to be a clustering of mutations that caused a more severe clinical phenotype towards the 3-prime end of the gene. The absence of large deletions, frameshift mutations, and nonsense mutations was considered consistent with the notion that a total lack of G6PD activity would be lethal.

Miwa and Fujii (1996) listed the mutations responsible for about 78 G6PD variants.

Mason (1996) reviewed information on the G6PD enzyme and on mutations in the gene. A map of 515 amino acids showing the location of mutations, including double mutations, was provided.

Filosa et al. (1996) analyzed fractionated blood cells in 4 heterozygotes for the class 1 G6PD mutations G6PD Portici (305900.0008) and G6PD Bari (1187G-T). In erythroid, myeloid, and lymphoid cell lineages there was a significant excess of G6PD-normal cells, suggesting that a selective mechanism operates at the level of pluripotent blood stem cells. They concluded that their studies provided evidence that severe G6PD deficiency adversely affects the proliferation or survival of nucleated blood cells.

Liu et al. (1997) reported a method of determination of clonality using allele-specific PCR (ASPCR) to detect exonic polymorphisms in p55 (305360) and G6PD. They demonstrated a significant sex difference in allele frequencies in African Americans but not in Caucasians, and linkage disequilibrium for the p55 and G6PD alleles in Caucasians but not in African Americans.

Vulliamy et al. (1998) determined the causative mutation in 12 cases of G6PD deficiency associated with chronic nonspherocytic hemolytic anemia. In 11 cases, the mutation they found had previously been reported in unrelated individuals. These mutations comprised 7 different missense mutations and a 24-bp deletion, G6PD Nara (305900.0052), previously found in a Japanese boy. Repeated findings of the same mutations suggest that a limited number of amino acid changes can produce the chronic nonspherocytic hemolytic anemia phenotype and be compatible with normal development. They found 1 new mutation, G6PD Serres (305900.0051).

Cappadoro et al. (1998) presented evidence suggesting that early phagocytosis of G6PD-deficient erythrocytes parasitized by Plasmodium falciparum may explain malaria protection in G6PD deficiency.

Kwok et al. (2002) described a Web-accessible database of G6PD mutations. The relational database integrates up-to-date mutational and structural data from various databanks with biochemically characterized variants and their associated phenotypes obtained from published literature and a Favism website.

Barisic et al. (2005) identified 5 different mutations in the G6PD gene in 24 unrelated males with G6PD deficiency from the Dalmatian region of southern Croatia. The variants included Cosenza (305900.0059) (37.5% of patients), Mediterranean (305900.0006) (16.6%), Seattle (12.5%), Union (12.5%), Cassano (4.2%), and a novel variant, termed G6PD Split (305900.0059) (4.2%). The variants in 3 patients (12.5%) were uncharacterized.

Ninokata et al. (2006) identified G6PD deficiency in 9.8% of males and 10.4% of females among 345 healthy adults on Phuket island in southern Thailand. Although none of the individuals had molecular evidence of malaria infection, the findings suggested that malaria endemics had occurred in the past and that G6PD deficiency has been maintained as an advantageous genetic trait in this population. At least 5 different G6PD variants were identified, suggesting that several Asian ethnic groups, such as Burmese, Laotian, Cambodian, Thai, and Chinese, participated in establishing the current ethnic identity of the population of Phuket.

Jiang et al. (2006) identified 14 different mutations in the G6PD gene among 1,004 G6PD-deficient Chinese individuals comprising 11 ethnic groups. The variants varied in frequency across the ethnic groups and correlated geographically with historical patterns of malaria. The variants were different from those reported in African, European, and Indian populations. The most common variants in the Chinese population were G6PD Kaiping (R463H; 305900.0029) and G6PD Canton (R459L; 305900.0021), accounting for over 60% of G6PD-deficient individuals, and Gaohe (H32R; 305900.0044). In vitro functional expression studies in E. coli showed significantly decreased enzyme activity for all 3 mutant proteins. All 3 variants showed decreased Km for G6P, but whereas the Canton and Kaiping variants had increased Km for NADP+, the Gaohe variant showed decreased Km for NADP+, likely reflecting compensation in the latter variant. Jiang et al. (2006) concluded that residues arg459 and arg463 play an important role in anchoring NADP+ to the catalytic domain of the enzyme.


Genotype/Phenotype Correlations

Miwa and Fujii (1996) stated that most of the class 1 G6PD variants associated with chronic hemolysis have the mutations surrounding either the substrate- or NADP-binding site.

Costa et al. (2000) pointed out that G6PD mutants causing class 1 variants (the most severe forms of the disease) cluster within exon 10, in a region that, at the protein level, is believed to be involved in dimerization. They identified a class 1 variant mapping to exon 8 (305900.0053).


Animal Model

Longo et al. (2002) crossed mouse chimeras from embryonic stem cells in which the G6pd gene had been targeted with normal females. First-generation G6pd heterozygotes born from this cross were essentially normal; their tissues demonstrated strong selection for cells with the targeted G6pd allele on the inactive X chromosome. When these first-generation heterozygous females were bred to normal males, only normal G6pd mice were born. There were 3 reasons for this: hemizygous G6pd male embryos' development was arrested from embryonic day 7.5, the time of onset of blood circulation, and they died by embryonic day 10.5; heterozygous G6pd females showed abnormalities from embryonic day 8.5, and died by embryonic day 11.5; and severe pathologic changes were present in the placenta of both G6pd hemizygous and heterozygous embryos. Thus, G6PD is not indispensable for early embryonic development; however, severe G6PD deficiency in the extraembryonic tissues (consequent on selective inactivation of the normal paternal G6PD allele) impairs the development of the placenta and causes death of the embryo. Most importantly, G6PD is indispensable for survival when the embryo is exposed to oxygen through its blood supply.

In ischemia-reperfusion experiments on isolated mouse hearts, Jain et al. (2004) demonstrated that G6pd is rapidly activated without a change in G6pd protein levels. G6pd -/- hearts had greatly impaired cardiac relaxation and contractile performance, associated with depletion of total glutathione stores and impaired generation of reduced glutathione, compared to wildtype hearts. Increased ischemia-reperfusion injury was reversed by antioxidant treatment but unaffected by supplementation of ribose stores. Jain et al. (2004) concluded that G6PD is an essential myocardial antioxidant enzyme, required for maintaining cellular glutathione levels and protecting against oxidative stress-induced cardiac dysfunction during ischemia-reperfusion.


History

Polymorphism at the G6PD locus has made it a useful X-chromosome marker, like the colorblindness and Xg blood group loci; close linkage of the colorblindness loci, the G6PD locus, and the hemophilia A locus (Adam et al., 1967; Boyer and Graham, 1965) has been demonstrated. Also, as a biochemical phenotype identifiable at the cellular level, G6PD variants have been useful in somatic cell genetics, permitting, for example, one of the critical proofs in man of the Lyon hypothesis (Davidson et al., 1963).

The relative stability of the X chromosome during evolution has been shown by the fact that the G6PD locus is X-borne also in a number of other species (Ohno, 1967). G6PD and HPRT are linked in the Chinese hamster (Rosenstraus and Chasin, 1975) and presumably are on the X chromosome as in man. By study of cell hybrids, Shows et al. (1976) found that HPRT and G6PD are closely linked in the Muntjac deer. Smith et al. (1976) found G6PD deficiency in a male Weimaraner dog, but were not able to do genetic studies. Alpha-GAL, HPRT, PGK and G6PD are X-linked in the rabbit, according to mouse-rabbit hybrid cell studies (Cianfriglia et al., 1979; Echard and Gillois, 1979). By comparable methods, Hors-Cayla et al. (1979) found them to be X-linked also in cattle. According to cell hybridization studies, HPRT, G6PD, and PGK are X-linked in the pig (Gellin et al., 1979) and in sheep (Saidi et al., 1979). Using pulsed field gel electrophoresis, Faust et al. (1992) demonstrated that, in the mouse, Gdx (312070), P3 (312090), and G6pd are physically linked to the X-linked visual pigment locus (Rsvp) within a maximal distance of 340 kb, while G6pd and f8 (300841) are approximately 900 kb apart.

Takizawa and Yoshida (1987) reported that the G6PD A+ gene has an A-to-G transition, resulting in the substitution of aspartic acid for asparagine as the 142nd amino acid from the N-terminus of the enzyme.

G6PD Hektoen is characterized by increased red cell enzyme activity. It is, therefore, a class 5 G6PD variant. It was first described by Dern et al. (1969). Yoshida (1970) thought that the variant peptide had replacement of histidine by tyrosine. Later, Yoshida (1996) was uncertain about this conclusion and stated that the basic defect remained to be identified.


ALLELIC VARIANTS ( 63 Selected Examples):

.0001 G6PD A+

G6PD, ASN126ASP
  
RCV000011073...

See Kirkman et al. (1964) and Yoshida et al. (1967). Vulliamy et al. (1988) found that the G6PD A variant is the same as 1 of the 2 variants identified in G6PD A- (305900.0002), i.e., asn126-to-asp. They noted that G6PD A, which is widely distributed in Africa, is not associated with deficiency of the enzyme.

Hirono and Beutler (1988) showed that a mutation responsible for the G6PD A- phenotype present in enzyme-deficient (300908) West African and American blacks occurred in a gene that produces the G6PD A+ phenotype. A substitution of guanine for adenine at nucleotide 376 (in exon 5) was found in all G6PD A+ and G6PD A- samples but in none of the G6PD B+ samples examined. Substitution of adenine for guanine at nucleotide 202 was found in 4 of 5 G6PD A- samples; this change is apparently responsible for the in vivo instability of the enzyme protein. Thus, the difference distinguishing the A and B forms of G6PD is the amino acid at residue 126 (see 305900.0002). Presumably as the result of alternative splicing, there is considerable heterogeneity among different G6PD cDNAs.

Both the variant A (with enzyme activity in the normal range, also called A) and the variant A- (associated with enzyme deficiency) have a frequency of about 0.2 in several African populations. Two restriction fragment length polymorphisms have also been found in people of African descent but not in other populations, whereas a silent mutation has been shown to be polymorphic in Mediterranean, Middle Eastern, African, and Indian populations. Vulliamy et al. (1991) reported 2 additional polymorphisms detected by sequence analysis, one in intron 7 and one in intron 8. Analysis of 54 African males for the 7 polymorphic sites clustered within 3 kb of the G6PD gene showed only 7 of the 128 possible haplotypes, thus indicating marked linkage disequilibrium. These data enabled Vulliamy et al. (1991) to suggest an evolutionary pathway for the different mutations, with only a single ambiguity. The mutation underlying the A variant is the most ancient and the mutation underlying the A- variant is the most recent. Since it seemed reasonable that the A- allele is subject to positive selection by malaria, whereas the other alleles are neutral, Vulliamy et al. (1991) suggested that G6PD may lend itself to the analysis of the role of random genetic drift and selection in determining allele frequencies within a single genetic locus in human populations.


.0002 G6PD A-

G6PD MATERA
G6PD BETICA
G6PD CASTILLA
G6PD DISTRITO FEDERAL
G6PD TEPIC
G6PD, VAL68MET, ASN126ASP
  
RCV000011073...

Babalola et al. (1976) predicted that the A- mutation may have occurred in an individual carrying the A+ mutation. A black individual with the G6PD A- phenotype but no mutation at nucleotide 202 suggested that this individual may have another mutation that caused instability and thus deficiency of the enzyme. Yoshida and Takizawa (1988) presented evidence that the A- gene evolved by stepwise mutations through the A+ gene.

Vulliamy et al. (1988) cloned and sequenced 7 mutant G6PD alleles. A single point mutation in the African variant G6PD A does not result in deficiency of the enzyme. The other 6 mutants, including G6PD A-, were all associated with enzyme deficiency. Two different point mutations were found in G6PD A-, 1 of which was the same as that in G6PD A. See Yoshida et al. (1967). Hirono and Beutler (1988) demonstrated a substitution of methionine for valine at position 68 resulting from a G-to-A change at nucleotide 202 (in exon 4). The in vivo instability of the enzyme is the result of this change. The gene also has the change at amino acid 126 characteristic of G6PD A. See Vulliamy et al. (1988).

Beutler et al. (1989) performed haplotyping with 4 polymorphic restriction sites in the G6PD locus in DNA samples from 29 males with the G6PD A- phenotype and 14 males with a G6PD B phenotype. All G6PD A- subjects with the G6PD A- (202A/376G) genotype, regardless of population origin, shared identical haplotypes. The 5 populations screened were black (16), Puerto Rican (2), Mexican (2), white US (1), and Spanish (3). One G6PD A- male was of the 376G/680T genotype and 2 were of the 376G/968C genotype. One of the restriction sites is uncommon in the populations studied; thus, Beutler and Kuhl (1990) considered it likely that the G6PD A- mutation at nucleotide 202 arose relatively recently and in a single person.

Calabro et al. (1990) found this mutation, regarded as characteristically African, in an unselected sample of 1,524 schoolboys of the province of Matera in Southern Italy.

Beutler et al. (1991) found that 3 previously reported electrophoretically fast Mexican G6PD variants--G6PD Distrito Federal (Lisker et al., 1981), G6PD Tepic (Lisker et al., 1985), and G6PD Castilla (Lisker et al., 1977)--all showed the changes characteristic of G6PD A- (202A/376G) and had the haplotype characteristic of G6PD A- in Africa. G6PD Betica (Vives-Corrons and Pujades, 1982; Vives-Corrons et al., 1980), which is frequent in Spain, also had the same characteristics. Since the PvuII+ genotype is rare in Europe, the G6PD Betica mutation was presumably imported from Africa.

Hirono and Beutler (1988) found 2 other mutations that produced the G6PD A- phenotype: arg227-to-leu and leu323-to-pro. In both cases the mutations existed on the G6PD A background, i.e., the asn126-to-asp substitution.

Town et al. (1992) demonstrated that both the val68-to-met and the asn126-to-asp mutations found in G6PD A- are necessary to produce the G6PD-deficient phenotype (rather than the val68-to-met mutation having happened to arise in an A+ gene in the first instance). They approached the question by introducing G6PD B, A, A-, and G6PD val68-to-met in a bacterial expression system and analyzing their biochemical properties. With each of the 2 mutations alone, they found a slight decrease in both the specific activity and the yield of enzyme protein when compared to G6PD B. When both mutations were introduced together, there was a roughly additive effect on specific activity, but a much more drastic effect on enzyme yield which was reduced to 4% of normal. They inferred that the coexistence of the 2 mutations acted synergistically in causing instability of the enzyme. This would explain why a B- phenotype has only very rarely been observed. (Comparable results were produced when the replacement gln119-to-glu was combined with val68 to met.)

G6PD A- is the most common polymorphic variant associated with deficiency of G6PD in African populations, accounting for 20 to 40% of the affected population in western and central Africa; the most common nondeficient polymorphic variant in Africa is G6PD A. The G6PD A- mutation at position 68 alone has not been detected in any variant; this, together with further haplotyping analyses, led Vulliamy et al. (1992) to suggest that the nondeficient single mutant G6PD A is more ancient than the deficient double mutant G6PD A-.

Gomez-Gallego et al. (2000) performed structural studies on the doubly mutant G6PD A-. The changes they observed did not affect the active site of the mutant protein, since its spatial position was unmodified. The result of the structural changes was a loss of folding determinants, leading to a protein with decreased intracellular stability. Gomez-Gallego et al. (2000) suggested that the resultant protein was the cause of the enzyme deficiency in the red blood cell, which is unable to perform de novo protein synthesis.


.0003 G6PD CHATHAM

G6PD, ALA335THR
  
RCV000011081...

Substitution of adenine for guanine at nucleotide 1003 leads to substitution of alanine by threonine at amino acid position 335 (Vulliamy et al., 1988). This mutation has been found in 2 unrelated Asian Indians and in a man from Syria and may be polymorphic. It causes class 2 enzyme derangement. No change in restriction sites has been found.

Mesbah-Namin et al. (2002) reported the first investigation of G6PD deficiency (300908) among the Mazandaranians of northern Iran. They analyzed the G6PD gene in 74 unrelated G6PD-deficient males with a history of favism. Molecular analysis revealed 3 different major polymorphic variants: G6PD Mediterranean (305900.0006) was found in 49 (66.2%), G6PD Chatham in 20 (27%), and G6PD Cosenza in 5 (6.75%) of the patients. The prevalence of G6PD Chatham in this Iranian population was the highest in the world. The distribution of the G6PD variants was more similar to that found in an Italian population than in other Middle Eastern countries.


.0004 G6PD ILESHA

G6PD, GLU156LYS
  
RCV000011082...

See Usanga et al. (1977) and Luzzatto et al. (1979). Substitution of adenine for guanine at base 466 (in exon 5) leads to replacement of glutamic acid by lysine (Vulliamy et al., 1988). This sporadic class 3 mutation is associated with loss of a HinfI site.


.0005 G6PD MAHIDOL

G6PD, GLY163SER
  
RCV000011085...

See Panich et al. (1972). A G-to-A change at base 487 (exon 6) leads to substitution of serine for glycine at amino acid 163 (Vulliamy, 1989). This mutation is polymorphic in Southeast Asia, causes class 2 enzyme derangement, and is associated with a new AluI site (Vulliamy et al., 1989). The same mutation was identified by Tang et al. (1992) in a Taiwanese in Taiwan.

Matsuoka et al. (2004) found that 11% of blood samples from persons in remote areas of Myanmar (former Burma) indicated G6PD deficiency. Taken together with data from a previous report (Iwai et al., 2001), these findings indicated that 91.3% of G6PD variants were G6PD Mahidol. The findings suggested that the Myanmar population is derived from homogeneous ancestries different from those of Thai, Malaysian, and Indonesian populations.

Louicharoen et al. (2009) investigated the effect of the G6PD-Mahidol 487A variant on human survival related to P. vivax and P. falciparum malaria in Southeast Asia. They showed that strong and recent positive selection has targeted the Mahidol variant over the past 1,500 years. The authors found that the G6PD-Mahidol variant reduces vivax, but not falciparum, parasite density in humans, which indicates that P. vivax has been a driving force behind the strong selective advantage conferred by this mutation.


.0006 G6PD MEDITERRANEAN

G6PD SASSARI
G6PD CAGLIARI
G6PD, SER188PHE
  
RCV000011086...

See Kirkman et al. (1964), Ben-Bassat and Ben-Ishay (1969), Lenzerini et al. (1969), Testa et al. (1980), and Morelli et al. (1984). A change from cytosine to thymine at base position 563 (in exon 6) causes a change from serine to phenylalanine in amino acid position 188 (Vulliamy et al., 1988). De Vita et al. (1989) found that G6PD Mediterranean, G6PD Sassari, and G6PD Cagliari have the same mutational change, resulting from a TCC-to-TTC mutation in exon 6. There is a second silent mutation of TAC-to-TAT at codon 437 in exon 11 (C-to-T at nucleotide 1311; see 305900.0018); both codons code for tyrosine. This mutation is a polymorphism, causes class 2 abnormality, and creates a new MboII site.

Beutler and Kuhl (1990) studied the distribution of the nucleotide polymorphism C1311T in diverse populations. Only 1 of 22 male subjects from Mediterranean countries who had the G6PD Mediterranean-563T genotype had a C at nucleotide 1311, which is the more frequent finding in this group. In contrast, both G6PD Mediterranean-563T males from the Indian subcontinent had the usual C at nucleotide 1311. Beutler and Kuhl (1990) interpreted these findings as suggesting that the same mutation at nucleotide 563 arose independently in Europe and in Asia.

Similar studies were done by Kurdi-Haidar et al. (1990) in 21 unrelated individuals with G6PD Mediterranean from Saudi Arabia, Iraq, Iran, Jordan, Lebanon, and Israel. All but 1 had the 563 mutation, and, of these, all but 1 had the C-to-T change at nucleotide 1311. Among another 24 unrelated Middle Eastern persons with normal G6PD activity, 4 had the silent mutation at position 1311 in the absence of the deficiency mutation at position 563. Kurdi-Haidar et al. (1990) concluded that most Middle Eastern subjects with the G6PD Mediterranean phenotype have the same mutation as that found in Italy; that the silent mutation is an independent polymorphism in the Middle East, with a frequency of about 0.13; and that the mutation leading to G6PD Mediterranean deficiency probably arose on a chromosome that already carried the silent mutation.

In Nepal, Matsuoka et al. (2003) tested 300 males for G6PD deficiency and identified 2 (0.67%) who were G6PD deficient. Compared with normal controls, G6PD activity was 12% and 26%, respectively. Both subjects had the 563C-T substitution of G6PD Mediterranean (ser188 to phe), and both had the silent 1311C-T change. A similar second change has been described in persons living in Mediterranean countries and Middle East countries. However, the form of G6PD Mediterranean found in India and Pakistan has no replacement at nucleotide 1311. Thus, the 2 subjects in Kathmandu, Nepal, would be closer to people in Middle East countries than people in India.

Corcoran et al. (1992) described a G6PD mutant biochemically indistinguishable from the common variety due to a C-to-T mutation at nucleotide 563. Instead, a C-to-T transition was found at nucleotide 592 in exon 6, changing an arginine residue to a cysteine residue only 10 amino acids downstream from the Mediterranean mutation. The new variant was named G6PD Coimbra (305900.0031).

Kaplan et al. (1997) presented data suggesting that the coexistence of Mediterranean type G6PD deficiency with the TA insertion polymorphism of the promoter of the UGT1A1 gene (191740.0011), which is associated with Gilbert syndrome (143500) in adults, is responsible for the development of neonatal hyperbilirubinemia. This is the most devastating clinical consequence of G6PD deficiency; it can be severe and result in kernicterus or even death. Kaplan et al. (1997) found that neither G6PD deficiency nor the polymorphism of UDP glucuronosyltransferase alone increased the incidence of neonatal hyperbilirubinemia, but in combination they did. The authors suggested that this gene interaction may serve as a paradigm of the interaction of benign genetic polymorphisms in the causation of disease.

Kaplan et al. (2001) reported 2 premature female neonates heterozygous for the G6PD Mediterranean mutation who presented with severe hyperbilirubinemia requiring exchange transfusions. Both had had normal G6PD biochemical screening tests.


.0007 G6PD METAPONTO

G6PD, ASP58ASN
  
RCV000011089...

Substitution of adenine for guanine at base 172 (exon IV) leads to a substitution of asparagine for aspartic acid at amino acid 58 (Vulliamy et al., 1988). The mutation was found in a sporadic, class 3 case, and no restriction site change was identified. See Calabro et al. (1990).


.0008 G6PD PORTICI

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, ARG393HIS
  
RCV000011090...

G6PD Portici has a G-to-A change at nucleotide 1178 of the G6PD gene, resulting in substitution of histidine for arginine at residue 393 (Filosa, 1989). The mutation was found in a sporadic case of class 1 deficiency (300908) and is not associated with an identified restriction site. In the full report, Filosa et al. (1992) described the kinetic characteristics of this G6PD variant (Portici) which was associated with chronic nonspherocytic hemolytic anemia.


.0009 G6PD SANTIAGO DE CUBA

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, GLY447ARG
  
RCV000011091...

Substitution of adenine for guanine as base number 1339 (exon 11) leads to substitution of arginine for glycine at amino acid position 447 (Vulliamy et al., 1988). This variant is associated with severe chronic hemolytic anemia (class 1; 300908). It was found in a sporadic case. A new PstI site was created, and this was used to show that it was a new mutation.


.0010 G6PD SEATTLE-LIKE

G6PD MODENA
G6PD, ASP282HIS
  
RCV000011092...

See Lenzerini et al. (1969) and Rattazzi et al. (1969). De Vita et al. (1989) found that G6PD Seattle-like, which produces a relatively mild phenotype, has substitution of histidine for aspartic acid at amino acid 282, resulting from a GAT-to-CAT change in exon 8. Cappellini et al. (1994) found the same variant in an Italian man from the Po delta and designated it G6PD Modena before finding that it had the same mutation as that in G6PD Seattle-like. They stated that the G-to-C transition was at nucleotide 844 in exon 8.


.0011 G6PD HARILAOU

G6PD, PHE216LEU
  
RCV000011094...

Town et al. (1990) described G6PD Harilaou in a Greek boy with severe hemolytic anemia. Poggi (1989) found a T-to-G change at nucleotide 648 that leads to substitution of leucine for phenylalanine at residue 216.


.0012 G6PD IOWA

G6PD IOWA CITY
G6PD SPRINGFIELD
G6PD WALTER REED
G6PD, LYS386GLU
  
RCV000011096...

See Beutler et al. (1986). Hirono et al. (1989) demonstrated an A-to-G substitution at nucleotide 1156, resulting in substitution of glutamic acid for lysine at amino acid 386. This variant G6PD, as well as G6PD Beverly Hills, Tomah, Riverside, and some others, is unstable in the presence of 10 microM NADP+ (where normal G6PD is stable) but is reactivated by 200 microM NADP+. G6PD Tomah, Iowa and Beverly Hills have amino acid substitutions at positions 385, 386, and 387, respectively; G6PD Riverside, with a substitution at position 410, shows weak reactivation by NADP+. These observations, together with the fact that these amino acids are highly conserved, led Hirono et al. (1989) to propose that they are in the region of the molecule involved in NADP+ binding.


.0013 G6PD BEVERLY HILLS

G6PD, ARG387HIS
  
RCV000011100...

Hirono et al. (1989) demonstrated a G-to-A mutation at nucleotide 1160, causing substitution of histidine for arginine-387. The mutation destroyed an HhaI site.


.0014 G6PD TOMAH

G6PD, CYS385ARG
  
RCV000011101...

Hirono et al. (1989) demonstrated a T-to-C transition at nucleotide 1153, causing substitution of arginine for cysteine-385. The mutation created an Fnu4HI restriction site, which was used to confirm the mutation.


.0015 G6PD RIVERSIDE

G6PD, GLY410CYS
  
RCV000011102...

Hirono et al. (1989) demonstrated a G-to-T mutation at nucleotide 1228 that caused a change of glycine to cysteine at amino acid 410. The fact that the mutation destroyed an NciI restriction site was used to confirm the mutation.


.0016 G6PD MONTALBANO

G6PD, ARG285HIS
  
RCV000011080...

Viglietto et al. (1990) found a new variant with nearly normal properties, due to a G-to-A transition that caused an arginine-to-histidine substitution at position 285. See Calabro et al. (1990).


.0017 G6PD RFLP

G6PD, NT1116, G-A
  
RCV000011083...

D'Urso et al. (1988) found a silent G-to-A change at nucleotide 1116 (in exon 10), generating a PstI site.


.0018 G6PD RFLP

G6PD, NT1311, C-T
  
RCV000011084...

De Vita et al. (1989) found a silent C-to-T change at nucleotide 1311 (in exon 11).


.0019 G6PD RFLP

G6PD, EX6, -60, C-G
  
RCV000011095...

Yoshida et al. (1988) found a RFLP resulting from a substitution in intron 5, creating a PvuII site. The probable change was C to G at a position 60 nucleotides upstream from exon 6 (Luzzatto, 1990).


.0020 G6PD ANDALUS

G6PD, ARG454HIS
  
RCV000011103...

Vives-Corrons et al. (1990) studied a G6PD variant resembling G6PD Mediterranean kinetically but with a slightly rapid electrophoretic mobility. They demonstrated a G-to-A transition at nucleotide 1361, producing an arg-to-his substitution.


.0021 G6PD CANTON

G6PD GIFU
G6PD AGRIGENTO
G6PD TAIWAN-HAKKA
G6PD, ARG459LEU
  
RCV000011104...

G6PD Canton is one of the most common deficient variants in Orientals, reaching a gene frequency of 1.7% in southern China (McCurdy et al., 1966). Stevens et al. (1990) demonstrated that codon 459 in G6PD-B is changed from CGT(arginine) to CTT(leucine). The G-to-T change occurs at nucleotide 1376. Tang et al. (1992) found this mutation in 3 Taiwanese and 1 Hakkanese in Taiwan. They pointed out that the same mutation occurs in 3 other Chinese G6PD variants in Guangdong, China: Taiwan-Hakka (McCurdy et al., 1970), Gifu (Fujii et al., 1984), and Agrigento (Sansone et al., 1975). The G6PD Gifu variant was discovered in a 9-year-old Japanese male with chronic hemolysis and hemolytic crises after upper respiratory infections (Fujii et al., 1984). Enzyme activity was 2.9% of normal. The patient's G6PD showed increased utilization of substrate analog, deamino-NADP, and thermal instability.


.0022 G6PD PUERTO LIMON

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, GLU398LYS
  
RCV000011108...

Beutler et al. (1991) found a G-to-A transition at nucleotide 1192 causing a substitution of the amino acid lysine for the normal glutamic acid at position 398. This aberrant G6PD associated with nonspherocytic hemolytic anemia (300908) was described by Elizondo et al. (1982).


.0023 G6PD SANTAMARIA

G6PD, ASP181VAL, ASN126ASP
  
RCV000011073...

Beutler et al. (1991) found an A-to-T mutation at nucleotide 542 resulting in an asp-to-val substitution at amino acid 181. The subjects were white with 'some evidence of hemolysis' in one but none in the other. This aberrant G6PD, described by Saenz et al. (1984) in 2 unrelated subjects from Costa Rica, is 1 of 4 polymorphic variants that have 2 point mutations. One of these point mutations in each case is 376A-G (asn126asp), the change characteristic of the nondeficient polymorphic variant G6PD A+ (305900.0001).


.0024 G6PD GASTONIA

G6PD MARION
G6PD MINNESOTA
ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, VAL213LEU
  
RCV000011110...

Beutler et al. (1991) found that although the patients from whom this variant G6PD was derived were unrelated, all had a G-to-T mutation at nucleotide 637 in exon 6 leading to substitution of leucine for valine-213. The G6PD variants called Gastonia, Marion, and Minnesota were all from patients with nonspherocytic hemolytic anemia (300908).


.0025 G6PD NASHVILLE

G6PD ANAHEIM
ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, ARG393HIS
   RCV000011090...

In 2 unrelated patients with nonspherocytic hemolytic anemia (300908), Beutler et al. (1991) found a G-to-A mutation at nucleotide 1178 in exon 10 producing substitution of histidine for arginine-393.


.0026 G6PD VIANGCHAN

G6PD JAMMU
G6PD, VAL291MET
  
RCV000011116...

See Poon et al. (1988). Beutler (1991) reported a G-to-A mutation at nucleotide 871, resulting in substitution of methionine for valine-291. The variant belonged to WHO class 2.

Louicharoen and Nuchprayoon (2005) and Matsuoka et al. (2005) indicated that G6PD Viangchan is the most common mutation in the Cambodian population, similar to Thai and Laotian populations, suggesting a common ancestry for people from these 3 countries. Matsuoka et al. (2005) found that G6PD Viangchan was linked in 8 cases with a 1311C-T transition (305900.0018) in exon 11 and a T-to-C substitution in intron 11, 93 bp downstream of exon 11. The finding was in accordance with studies of G6PD Viangchan in Laos, Thailand, and Malaysia.


.0027 G6PD A-

G6PD, ARG227LEU
  
RCV000011118...

In subjects with the G6PD A- phenotype, Hirono and Beutler (1988) found substitution of leucine for arginine-227, resulting from a G-to-T mutation at nucleotide 680 (rather than the val68-to-met mutation as in the usual G6PD A-). The mutation existed on the G6PD A background (asn126 to asp).


.0028 G6PD A-

G6PD, LEU323PRO
  
RCV000011119...

In subjects with the G6PD A- phenotype, Hirono and Beutler (1988) found substitution of proline for leucine-323, resulting from a T-to-C mutation at nucleotide 968 (rather than the val68-to-met mutation as in the usual G6PD A-). The mutation existed on the G6PD A background (asn126-to-asp).


.0029 G6PD KAIPING

G6PD ANANT
G6PD DHON
G6PD PETRICH-LIKE
G6PD SAPPORO-LIKE
G6PD, ARG463HIS
  
RCV000011120...

Zuo et al. (1990) demonstrated substitution of histidine for arginine-463 resulting from a G-to-A mutation in nucleotide 1388. The G6PD was of the WHO class 2. The Chinese variant G6PD Kaiping was discovered by Du et al. (1988). The same mutation was found in G6PD Anant (Panich and Sungnate, 1973), Dhon (Panich and Na-Nakorn, 1980), Petrich-like (Shatskaya et al., 1980), and Sapporo-like (Fujii et al., 1981).


.0030 G6PD LOMA LINDA

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, ASN363LYS
  
RCV000011125...

In a patient with nonspherocytic hemolytic anemia (300908), Beutler et al. (1991) identified a C-to-A mutation at nucleotide 1089 in exon 10, producing substitution of asparagine-363 by lysine.


.0031 G6PD COIMBRA

G6PD, ARG198CYS
  
RCV000011126...

In the son of a Portuguese woman who had suffered an attack of favism, Corcoran et al. (1992) identified a G6PD mutant with the chemical properties of the Mediterranean type (305900.0006). However, at the DNA level, they demonstrated that the mutation was a C-to-T transition 29 nucleotides downstream from the Mediterranean mutation, resulting in substitution of cysteine for arginine 10 amino acids downstream from the Mediterranean change. The same mutation was found in a patient in southern Italy. The new variant was called G6PD Coimbra.

In 3 individuals with G6PD deficiency from tribal groups in southern India, Chalvam et al. (2008) identified the Coimbra variant and stated that the mutation had a frequency of 7.5% in this population.


.0032 CHRONIC GRANULOMA AND HEMOLYTIC ANEMIA

G6PD, SER106CYS, ARG182TRP, ARG198CYS
  
RCV000011126...

Gray et al. (1973) described a unique G6PD variant in a patient with chronic granuloma and hemolytic anemia. G6PD activity was undetectable not only in the patient's red blood cells but also in leukocytes and fibroblasts, and an immunologically crossreacting material was undetectable in these tissues. This is the only variant observed with no measurable activity and lack of crossreacting material, satisfying the definition for a 'null' variant. Maeda et al. (1992) found that the mRNA content and the size of mRNA were normal in the patient's lymphoblastoid cells (maintained as GM7254 in the Coriell repository in Camden, New Jersey). Western blot hybridization indicated that the patient's cells did not produce crossreacting material. Three nucleotide base changes were found in variant cDNA: a C-to-G transversion at nucleotide 317 (counting from adenine of the initiation codon), which should cause a ser-to-cys substitution at the 106th position (counting from the initiation met); a C-to-T transition at nucleotide 544, producing an arg-to-trp substitution at the 182nd position; and a C-to-T transition at nucleotide 592, resulting in an arg-to-cys substitution at the 198th position of the protein. No deletions or frameshift mutations were found, and no nucleotide change was detected in the extended 5-prime region which included the most distal cap site. When the variant cDNA was expressed in E. coli, the G6PD activity was about 2% of normal and crossreacting material was undetectable. However, when the variant mRNA was expressed in the in vitro translation system of rabbit reticulocytes, the variant protein was produced. The results suggested that extremely rapid in vivo degradation or precipitation of the variant enzyme induced by the 3 amino acid substitutions could be the major cause of the molecular deficiency.


.0033 G6PD TAIWAN-HAKKA 2

G6PD, ASN165ASP
  
RCV000011128...

Tang et al. (1992) identified an A-to-G transition at nucleotide 493 resulting in an asn165-to-asp amino acid substitution in the G6PD protein. The biochemical features of the mutation were not characterized. This mutation has only been reported in Chinese.

The Chinese population of Taiwan is divided into 4 groups: Taiwanese, mainland Chinese, Hakkanese, and Aborigines. The Taiwanese, the largest group, are descendants from emigrants who left mainland China during the 17th to 19th centuries. Most were from Fuchien Province on the southeast coast of China. The second largest population is mainland Chinese, who resided originally in many provinces throughout mainland China and migrated to Taiwan during the period 1948 to 1950. The third population is Hakkanese (Taiwan-Hakka), originally from Chung Yuan, who immigrated from the Kwangtung and Fuchien provinces on the southern coast of China and who came to Taiwan primarily during the 16th and 17th centuries. The native Taiwan Aborigines are a much smaller group, containing at least 9 distinct tribes whose ancestors are believed to have arrived in Taiwan from mainland Asia several thousand years ago. The frequency of G6PD deficiency varies from 4.52% in the Hakkanese to an average of 0.3% in most of the Aborigines. The Ami tribe of Aborigines shows a frequency of 3.5%, presumably a reflection of founder effect.


.0034 G6PD SANTIAGO

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, ARG198PRO
  
RCV000011129...

In a Chilean patient with nonspherocytic hemolytic anemia (300908), Beutler et al. (1992) identified a G-to-C transversion at nucleotide 593 leading to an arg198-to-pro substitution. They suggested G6PD Santiago as the designation. (G6PD Santiago de Cuba is a different mutation; see 305900.0009.)


.0035 G6PD MEXICO CITY

G6PD, ARG227GLN
  
RCV000011131...

In a Mexican individual with no clinical features attributable to the G6PD variant, Beutler et al. (1992) described a G-to-A transition at nucleotide 680 leading to an arg227-to-gln substitution. They suggested the designation G6PD Mexico City. (There is a G6PD Mexico; see under 305900.9999.) Nucleotide 680 is the same base that is altered from G-to-T in one type of G6PD A- (arg227-to-leu).


.0036 G6PD IERAPETRA

G6PD, PRO353SER
  
RCV000011115...

In a Greek person with no clinical abnormalities that could be related to the G6PD variant, Beutler et al. (1992) identified a C-to-T transition at nucleotide 1057 resulting in a pro353-to-ser substitution.


.0037 G6PD GUADALAJARA

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, ARG387CYS
  
RCV000011132...

See Vaca et al. (1982). In a Mexican patient with nonspherocytic hemolytic anemia (300908), Beutler et al. (1992) identified an arg387-to-cys substitution resulting from a C-to-T transition at nucleotide 1159.


.0038 G6PD ALHAMBRA

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, VAL394LEU
  
RCV000011134...

See Beutler and Rosen (1970). Beutler et al. (1992) indicated that the mutation in this G6PD variant found in a US white patient with nonspherocytic hemolytic anemia (300908) involved a G-to-C transversion at nucleotide 1180 resulting in a val394-to-leu substitution.


.0039 G6PD JAPAN

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, GLY410ASP
  
RCV000011136...

In a Japanese patient with nonspherocytic hemolytic anemia (300908), Beutler et al. (1992) identified a G-to-A transition at nucleotide 1229 resulting in a gly410-to-asp substitution.


.0040 G6PD PAWNEE

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, ARG439PRO
  
RCV000011138...

In a US white patient with nonspherocytic hemolytic anemia (300908), Beutler et al. (1992) identified a G-to-C transition at nucleotide 1316 leading to an arg439-to-pro substitution.


.0041 G6PD SUNDERLAND

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, ILE35DEL
  
RCV000011140...

Using a PCR-based technique, MacDonald et al. (1991) determined the nucleotide sequence of the entire coding region of the G6PD gene from a person with severe red cell G6PD deficiency and chronic hemolytic anemia (300908). The only abnormality found was a 3-bp deletion in exon 2, which predicted the loss of 1 of 2 adjacent isoleucine residues (amino acid 35 or 36), just upstream of the methionine residue called 'junctional' by Kanno et al. (1989). This part of exon 2 lies in a region that was thought by Kanno et al. (1989) to be encoded by a gene on chromosome 6, an idea subsequently disproved. The observations of MacDonald et al. (1991) demonstrated that a mutation in this X-linked amino-terminal region of G6PD caused deficiency in red cells. The deletion was within a 3-fold CAT repeat and had presumably resulted from misalignment at meiosis, with conservation of the reading frame.


.0042 G6PD KERALA-KALYAN

G6PD KERALA
G6PD KALYAN
G6PD, GLU317LYS
  
RCV000011142...

G6PD Kerala (Azevedo et al., 1968) and G6PD Kalyan (Ishwad and Naik, 1984), 2 variants discovered in India, were thought to be distinct on the basis of their biochemical properties. Ahluwalia et al. (1992) demonstrated that the molecular defect is identical. Both have a glu317-to-lys mutation which causes a loss of 2 negative charges; this is in keeping with the very slow electrophoretic mobility of G6PD Kerala-Kalyan. Both are accompanied by only mild enzyme deficiency. In both, the mutation is a C-to-T transition in the CpG dinucleotide. The mutations were found in 2 populations that are entirely distinct linguistically and culturally with no known historical links. However, in light of the traditional occupation of the Koli tribal group inhabiting the Kalyan district of Bombay, namely, marine fishing, victims of bad weather may have found their way to distant places where they were forced to live for some period, thus creating the possibility of gene flow.


.0043 G6PD AURES

G6PD, ILE48THR
  
RCV000011145...

In an Algerian boy who presented to the hospital with acute hemolytic anemia associated with 7 to 10% of G6PD residual activity, Nafa et al. (1993) identified a T-to-C transition at nucleotide 143 converting codon 48 from ATC (ile) to ACC (thr). The mutation was associated with favism.

In Saudi Arabia, Niazi et al. (1996) described G6PD Aures in 7 of 20 children (35%) with severe G6PD deficiency and in a 16-year-old boy with a history of passing dark urine after eating fava beans at the age of 5 years. Of the 20 children, 12 were positive for G6PD Mediterranean (305900.0006), and the mutation in 1 child remained unidentified.


.0044 G6PD GAOHE

G6PD, HIS32ARG
  
RCV000011146...

This G6PD variant was described by Du et al. (1985). Its biochemical characterization was reviewed by Chiu et al. (1993), who demonstrated that the mutant is frequent in Chinese and consists of a change in cDNA nucleotide 95 from A to G (his to arg).


.0045 G6PD QUING YUAN

G6PD, GLY131VAL
  
RCV000011147...

In an analysis of the molecular defect in 43 G6PD-deficient Chinese, Chiu et al. (1993) found 3 with a G-to-T transversion in cDNA nucleotide 392 (exon 5) resulting in a gly-to-val substitution. They reviewed the biochemical characteristics of this previously unidentified variant.


.0046 G6PD MAHIDOL-LIKE

G6PD, LEU342PHE
  
RCV000011148...

In a study of the molecular defect in 43 G6PD-deficient Chinese, Chiu et al. (1993) identified a 'new' variant due to a C-to-T transition at cDNA nucleotide 1024 resulting in a leu-to-phe substitution. Chiu et al. (1993) listed the biochemical characteristics of G6PD Mahidol-like.


.0047 G6PD ORISSA

G6PD, ALA44GLY
  
RCV000011149...

To determine the extent of heterogeneity of G6PD in India, Kaeda et al. (1995) studied several different Indian populations by screening for G6PD deficiency, followed by molecular analysis of deficient alleles. The frequency of G6PD deficiency varied between 3% and 15% in different tribal and urban groups. Remarkably, a previously unreported deficient variant, G6PD Orissa (ala44-to-gly), was found to be responsible for most of the G6PD deficiency in tribal Indian populations but was not found in urban populations where most of the G6PD deficiency was due to the G6PD Mediterranean (ser188-to-phe) variant (305900.0006). The distribution of G6PD alleles in India is reminiscent of the situation found with beta-globin (141900), as reviewed by Nagel and Ranney (1990). In that case, sickle cell anemia is almost entirely restricted to the tribal groups, whereas urban populations have a predominance of beta-thalassemia mutations. Kaeda et al. (1995) noted that the Km(NADP) of G6PD Orissa was 5-fold higher than that of the normal enzyme. This was thought to be due to the fact that the alanine residue that is replaced by glycine is part of a putative coenzyme-binding site. Surprisingly, the enzyme appeared to the authors to be more stable than normal G6PD, whereas most deficient variants have lowered stability.


.0048 G6PD NANKANG

G6PD, PHE173LEU
  
RCV000011150...

In a Chinese newborn with neonatal jaundice, Chen et al. (1996) identified a novel G6PD mutation, G6PD NanKang, caused by a T-to-C transition at nucleotide 517, producing a phe173leu substitution in the G6PD protein.


.0049 G6PD MALAGA

G6PD, ASP181VAL
  
RCV000011109...

In a study of G6PD-deficient patients who presented with clinical favism in Spain, Vulliamy et al. (1996) found a new polymorphic variant they called G6PD Malaga, whose only abnormality was an A-to-T transversion at nucleotide 542 resulting in an asp181-to-val amino acid substitution. This was the same mutation previously found in association with the mutation of G6PD A-, namely asn126asp (305900.0001) in the double mutant G6PD Santamaria (305900.0023). G6PD Malaga was associated with enzyme deficiency class 3, and the enzymic properties of G6PD Malaga and G6PD Santamaria were quite similar. Vulliamy et al. (1996) speculated that G6PD Santamaria might have been produced by recombination between G6PDA and G6PD Malaga; however, haplotype analysis, including the use of a new silent polymorphism, suggested that the same 542A-T mutation had taken place independently in a G6PD B gene to give G6PD Malaga and in a G6PD A gene to give G6PD Santamaria.


.0050 G6PD NEAPOLIS

G6PD, PRO467ARG
  
RCV000011152...

In a study of 31 unrelated G6PD-deficient males in the Campania region of Southern Italy, Alfinito et al. (1997) found 9 different G6PD variants, 8 of which had already been described. The new variant, G6PD Neapolis, was found to have a pro467-to-arg substitution in the G6PD protein.


.0051 G6PD SERRES

ANEMIA, NONSPEHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, ALA361VAL
  
RCV000011153...

In a study of the causative mutation in 12 cases of G6PD deficiency associated with chronic nonspherocytic hemolytic anemia (300908), Vulliamy et al. (1998) found 1 patient to have a novel mutation, which they called G6PD Serres: a 1082C-T change, causing an ala361-to-val substitution in the dimer interface where most other severe G6PD mutations are found.


.0052 G6PD NARA

ANEMIA, NONSPEHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, 24-BP DEL, NT953
  
RCV000143789...

In a Japanese boy with severe G6PD deficiency (300908), Hirono et al. (1993) identified a 24-bp deletion (nucleotides 953-976) in exon 9 of the G6PD gene, which predicted an 8-amino acid deletion at residue 319.


.0053 G6PD AVEIRO

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, CYS269TYR
  
RCV000011155...

In a boy born in Aveiro, Portugal with severe chronic hemolytic anemia (300908) present from birth, Costa et al. (2000) found that the undetectable G6PD activity was caused by a G-to-A transition at nucleotide position 806 of the G6PD gene resulting in a cys269-to-tyr (C269Y) amino acid substitution. This mutation, which was designated G6PD Aveiro, was not detected in his mother or sister. By the age of 5 years, the patient had had 6 episodes of severe acute intravascular hemolysis that required hospitalization and erythrocyte transfusion. The spleen was palpable 6 cm below the left costal margin. Costa et al. (2000) pointed out that G6PD mutants causing class 1 variants (the most severe forms of the disease) cluster within exon 10, in a region that, at the protein level, is believed to be involved in dimerization. The mutation in this new class 1 variant maps to exon 8. Mutant and normal alleles were found in both hematopoietic and buccal cells, indicating mosaicism.


.0054 G6PD ASAHI

G6PD, VAL68MET
  
RCV000011075...

G6PD A- is a common G6PD variant among Africans that may cause acute hemolysis triggered by infections and certain drugs, as well as by fava beans. This class 3 phenotype can be caused by a combination of the common 376A-G (asn126 to asp) mutation and either of 3 additional mutations that include 202G-A (val68 to met); see 305900.0002. The missense mutation 376A-G (asn126 to asp) by itself causes an asymptomatic class 4 variant G6PD A with normal enzyme activity, whereas the other mutation, 202G-A, had not been found in humans by itself. Hirono et al. (2002) described an asymptomatic G6PD-deficient patient with the missense mutation 202G-A but not the 376A-G. This was a 3-year-old Japanese boy who was noted to have jaundice and anemia on admission to the Asahi General Hospital. This was the only mutation found and it must have arisen separately from those common in Africans, because the patient had none of the silent mutations closely linked to the African mutation, while he had an intronic single base deletion common in Mongoloids. Town et al. (1992) had found in an in vitro study using recombinant human G6PD mutants expressed in E. coli that 202G-A, as well as 376A-G, does not cause enzyme deficiency by itself, and the synergistic action of these 2 mutations is necessary to produce the class 3 phenotype of G6PD A-. Synergistic interaction was also supported by the fact that val68 and asn126 are closely located in a 3-dimensional model of human G6PD. The results of Hirono et al. (2002) seem inconsistent with the idea that 202G-A cannot produce acute hemolysis by itself.


.0055 G6PD REHOVOT

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, TYR322HIS
  
RCV000011158...

In 3 brothers and their carrier mother of Jewish Ethiopian descent, Iancovici-Kidon et al. (2000) found a T-to-C transition at nucleotide 964 in exon 9 of the G6PD gene, resulting in a tyr322-to-his (Y322H) mutation. All 3 sibs showed hereditary nonspherocytic hemolytic anemia (300908), but the severity of hemolysis and the transfusion requirement varied markedly. One brother had severe congenital neutropenia (SCN; 202700), a condition not previously described in association with G6PD deficiency. Levels of white blood cell G6PD activity of the 3 sibs was 0 to 5% of normal controls. Neutrophil oxidative and bactericidal activities were impaired in the brother with SCN, but were well preserved in the other 2 sibs.


.0056 G6PD AMSTERDAM

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, 3-BP DEL, 180TCT
  
RCV000066279

In a study of blood cells of 4 male patients from 2 unrelated families with nonspherocytic anemia (300908) and recurrent bacterial infections, van Bruggen et al. (2002) discovered that the activity of G6PD in red blood cells and in granulocytes was below detection level. Moreover, their granulocytes displayed a decreased respiratory burst upon activation. Sequencing of genomic DNA revealed a novel 3-bp (TCT) deletion in the G6PD gene, predicting the deletion of a leucine at position 61. The mutant G6PD protein was undetectable by Western blotting in red blood cells and granulocytes of these patients. In phytohemagglutinin-stimulated lymphocytes, the G6PD protein was present, but the amount of the protein was greatly diminished in the patients' cells. Purified mutant protein from an E. coli expression system showed decreased heat stability and decreased specific activity. Furthermore, van Bruggen et al. (2002) demonstrated that mRNA of the mutant G6PD was unstable, which may contribute to the severe G6PD deficiency observed in these patients. They proposed the name 'G6PD Amsterdam' for the new variant.

One family reported by van Bruggen et al. (2002) was Caucasian, the second was Hindustani. The Caucasian patient had an unremarkable medical history until he was admitted to the hospital at age 15 years with recurrent episodes of fever, jaundice, gastroenteritis, and coughing. He was found to have invasive disseminated aspergillosis (see 614079) in the lungs, brain, and soft tissues of the leg. Aspergillosis was successfully treated. Thereafter, hemoglobin level was normal but reticulocytosis was persistent. One of his brothers also had G6PD deficiency and presented with prolonged neonatal jaundice and episodes of acute hemolysis but had no known disposition to infections.

The Hindustani proband reported by van Bruggen et al. (2002) was healthy until the age of 3.5 years, when he was admitted to the hospital with pneumonia caused by Chromobacterium violaceum, an uncommon human pathogen that can cause serious infections in patients with neutrophil dysfunction. He was anemic. He responded well to chemotherapy, although the anemia persisted. With relapse he developed osteomyelitis but again responded to therapy.


.0057 G6PD ZURICH

G6PD, IVS10AS, A-G, -2
  
RCV000011165...

In a 33-year-old Swiss male with G6PD deficiency, designated G6PD Zurich, Efferth et al. (2004) identified a single nucleotide mutation that altered position -2 of intron 10 of the G6PD gene from the consensus A to G. The mutation resulted in alternative splicing that removed the first 9 nucleotides of exon 11, which code for amino acids asparagine, valine, and lysine at positions 430-432, respectively. Efferth et al. (2004) estimated that 400 million people worldwide are affected by G6PD deficiency, the most common hereditary enzymopathy, with some 140 known molecular G6PD defects. They pointed out that most mutations in the G6PD gene are missense mutations. To the best of their knowledge, there was only 1 missplicing mutation previously described: G6PD Varnsdorf (305900.0058), which is caused by destruction of the same obligate splice site as that destroyed in G6PD Zurich. In G6PD Varnsdorf, the invariant 3-prime AG dinucleotide has been deleted, whereas in G6PD Zurich, a point mutation has changed AG to GG. In both G6PD Zurich and G6PD Varnsdorf, the next available downstream consensus splice sequence is used, resulting in deletion of 3 amino acids. Efferth et al. (2004) suggested that it was no coincidence that the only 2 splicing mutations of G6PD identified to that time both affected the same splice site. Since null mutations of G6PD appear to be incompatible with life, a functional alternative splice site that does not cause a frameshift is required for viability. The 3-prime splice site of intron 10 offers this opportunity.


.0058 G6PD VARNSDORF

G6PD, IVS10AS, 2-BP DEL, AG, -2
  
RCV000011166...

.0059 G6PD COSENZA

G6PD, ARG459PRO
  
RCV000011167...

Calabro et al. (1993) identified a novel G6PD variant, which they called Cosenza, in patients with G6PD deficiency from the Calabria region of southern Italy. The arg459-to-pro (A459P) substitution results from a 1376G-C transversion. The mutant protein retains less than 10% enzyme activity and belongs to the group of severe disorders often associated with hemolysis.

Barisic et al. (2005) identified G6PD Cosenza in 9 (37.5%) of 24 unrelated patients with G6PD deficiency from the Dalmatian region of southern Croatia. Seven of the 9 patients had favism.


.0060 G6PD SPLIT

G6PD, PRO481ARG
  
RCV000011159...

In a male with G6PD deficiency from the Dalmatian region of southern Croatia, Barisic et al. (2005) identified a 1442C-G transversion in the G6PD gene, resulting in a pro481-to-arg (P481R) substitution. The mutant protein retained approximately 30% enzyme activity (class 3).


.0061 G6PD NAMORU

G6PD, HIS70TYR
  
RCV000011160...

Chalvam et al. (2007) identified a 208T-C transition in exon 4 of the G6PD gene, resulting in a his70-to-tyr (H70Y) substitution, as the basis of G6PD deficiency in Indian patients with the disorder. The H70Y mutation was detected in 28 (70.4%) of 40 affected Indian males from 3 tribal groups from the Nilgiri district of Tamil Nadu in southern India. The variant was termed G6PD Namoru.


.0062 G6PD NILGIRI

G6PD, ARG198HIS
  
RCV000011161...

In 4 individuals with G6PD deficiency from tribal groups of the Nilgiri district in southern India, Chalvam et al. (2008) identified a 593G-A transition in exon 6 of the G6PD gene, resulting in an arg198-to-his (R198H) substitution, which they designated G6PD Nilgiri. The authors stated that the mutation had a frequency of 10.0% in this population.


.9999 GLUCOSE-6-PHOSPHATE DEHYDROGENASE VARIANTS, MOLECULAR DEFECT UNKNOWN

G6PD VARIANTS, MOLECULAR DEFECT UNKNOWN

The following list of G6PD variants which have not been characterized at the molecular level is in alphabetic order. Quotation marks surround the name of each G6PD variant about which there is insufficient information for certainty of its uniqueness.

G6PD AACHEN. See Kahn et al. (1976).

G6PD AARAU. See Gahr et al. (1976).

G6PD 'ABEOKUTA'. See Usanga et al. (1977).

G6PD ABRAMI. See Kahn et al. (1975).

G6PD 'ADAME'. See Usanga et al. (1977).

G6PD ADANA. See Aksoy et al. (1987).

G6PD AKITA. See Miwa et al. (1978).

G6PD ALABAMA. Prchal et al. (1988) described a 6-year-old black boy who had transient hemolysis after a viral infection and was found to have mildly decreased red cell G6PD activity. The unusual finding was the presence of 2 G6PD bands in him and in his maternal grandfather despite normal XY karyotype. Two bands were seen only in reticulocytes. Prchal et al. (1988) postulated that there were 2 transcriptional products of the mutant G6PD gene, 1 of which had a short half-life and was detectable only in young red blood cells.

G6PD ALBUQUERQUE. See Beutler et al. (1968).

G6PD ALESSANDRIA. Similar to G6PD Alexandra. See Sansone et al. (1981).

G6PD ALEXANDRA. This was found in Australia in a male of Italian extraction who suffered severe neonatal jaundice following maternal ingestion of fava beans prenatally and postnatally. Retesting in adolescence showed milder expression of the enzyme defect (Harley et al., 1978).

G6PD ALGER. See Benabadji et al. (1978).

G6PD AMBOIN. See Chockkalingam et al. (1982).

G6PD AMMAN-1. See Karadsheh et al. (1986).

G6PD AMMAN-2. See Karadsheh et al. (1986).

G6PD ANGORAM. See Chockkalingam et al. (1982).

G6PD ANKARA. See Kahn et al. (1975).

G6PD ARLINGTON HEIGHTS. See Honig et al. (1979).

G6PD ASAHIKAWA. This variant was discovered in a 6-year-old Japanese boy with chronic hemolytic anemia and hemolytic crises after upper respiratory infections (Takizawa et al., 1984).

G6PD ASHDOD. See Ramot et al. (1969).

G6PD ATHENS. See Stamatoyannopoulos et al. (1967).

G6PD 'ATHENS-LIKE'. See Stamatoyannopoulos et al. (1971).

G6PD ATLANTA. See Beutler et al. (1976).

G6PD 'ATTICA'. See Rattazzi et al. (1969).

G6PD AVENCHES. See Pekrun et al. (1989).

G6PD 'AVVOCATA'. See Colonna-Romano et al. (1985).

G6PD AYUTTHAYA. See Panich (1980).

G6PD AZERBAIJAN. See Shatskaya et al. (1975).

G6PD B. The so-called normal, this form predominates in all populations greater than a few hundred (Yoshida et al., 1971).

G6PD 'BAGDAD'. See Geerdink et al. (1973).

G6PD BAKU. See Shatskaya et al. (1980).

G6PD 'BALCALI'. See Aksoy et al. (1987).

G6PD BALI. See Chockkalingam et al. (1982).

G6PD BALTIMORE-AUSTIN. See Porter et al. (1964) and Long et al. (1965).

G6PD BANGKOK. See Talalak and Beutler (1969).

G6PD BARBIERI. See Marks et al. (1962).

G6PD BARCELONA. See Vives-Corrons et al. (1982). This is one of the rare G6PD variants associated with granulocyte dysfunction and increased susceptibility to infections. Hemolysis in this form of chronic nonspherocytic hemolytic anemia is exaggerated by infection.

G6PD 'BASH-KUNGUT I AND II'. See Shatskaya et al. (1980).

G6PD 'BASH-KUNGUT IV'. See Shatskaya et al. (1980).

G6PD BAT-YAM. See Ramot et al. (1969).

G6PD BAUDELOCQUE. See Junien et al. (1974).

G6PD 'BEAUJON'. See Boivin and Galand (1968).

G6PD BEAUMONT. Mamlok et al. (1985) reported a new molecular variant associated with severe enzyme deficiency and chronic nonspherocytic hemolytic anemia. The characteristics were marked heat lability, a normal rate constant value for glucose-6-phosphate, a nearly normal pH activity curve, and increased use of 2-deoxyglucose-6-phosphate. Mamlok et al. (1987) described a fatal case of Chromobacterium violaceum sepsis in a 3-year-old boy with this variant. The child was an identical twin; the surviving twin subsequently had a severe episode of Campylobacter jejuni gastroenteritis. Patients with severe deficiency of G6PD and polymorphonuclear leukocytes have increased susceptibility to infections and abnormal phagocyte function that resembles that of patients with chronic granulomatous disease, but such had not hitherto been reported during the first decade of life. Infections with C. violaceum are rare; most of the 20 or so infections have occurred in Louisiana or Florida and have been associated with warm, stagnant water sources.

G6PD BENEVENTO. See McCurdy et al. (1973).

G6PD BERLIN. See Helge and Borner (1966).

G6PD BIDEIZ. See Krasnopolskaya et al. (1977).

G6PD BIELEFELD. See Gahr et al. (1977).

G6PD BIRMINGHAM. See Prchal et al. (1980).

G6PD BLIDA. See Benabadji et al. (1978).

G6PD BNEI BRAK. See Sidi et al. (1980).

G6PD BODENSEE. See Benohr et al. (1971).

G6PD BOGIA. See Chockkalingam and Board (1980).

G6PD BOLUO. See Du et al. (1988).

G6PD BOLUO-2. See Du et al. (1988).

G6PD BOSTON. See Necheles et al. (1971).

G6PD BUKITU. See Chockkalingam and Board (1980).

G6PD BUTANTAN. In Brazil, Stocco dos Santos et al. (1991) described a Gd(+) variant which was characterized by normal activity and electrophoretic mobility, increased Km, and increased activity for 2-deoxy-G6P. The variant, which they called G6PD Butantan, was present in 3, and perhaps a fourth, cousin; the 4 mothers were sisters. All 4 males had severe mental retardation, bilateral congenital hip luxation, and short stature. Five uncles of these males may have been affected. In this family, Stocco dos Santos et al. (2003) found linkage of the X-linked mental retardation syndrome (300434) to the pericentric region, Xp11.3-q21.1.

G6PD 'CAGLIARI II' (CAGLIARI-LIKE). See Frigerio et al. (1987) and Calabro et al. (1990).

G6PD 'CALTANISSETTA'. See Sansone et al. (1981) and Perroni et al. (1982).

G6PD 'CAMALDOLI'. See Colonna-Romano et al. (1985).

G6PD CAMPBELLPORE. See McCurdy et al. (1970).

G6PD CAMPERDOWN. Harley et al. (1978) found this variant in Australia in a boy of Maltese extraction in whom lamellar cataracts were found at age 4. The enzyme deficiency was detected in a screening of children of Mediterranean extraction with lamellar cataracts. The boy had no excessive hemolysis. Previous descriptions of cataracts were in patients with hemolytic anemia.

G6PD CAPETOWN. See Botha et al. (1969).

G6PD CARSWELL. See Siegel and Beutler (1971).

G6PD CASTILLA-LIKE. See Chockkalingam et al. (1982).

G6PD CAUJERI. See Gutierrez et al. (1987).

G6PD CENTRAL CITY. See Csepreghy et al. (1988).

G6PD CHAINAT. See Panich and Na-Nakorn (1980).

G6PD CHAO PHYA. See Panich (1980).

G6PD CHARLESTON. See Beutler et al. (1972).

G6PD CHIAPAS. See Lisker et al. (1978).

G6PD CHIBUTO. See Reys et al. (1970).

G6PD CHICAGO. See Kirkman et al. (1964) and Fairbanks et al. (1980). Fairbanks et al. (1980) demonstrated that G6PD Chicago and G6PD Cornell are the same variant; they had been described previously in different members of a single large kindred.

G6PD CHINESE. See Chan et al. (1972).

G6PD CIUDAD DE LA HABANA. See Gonzalez et al. (1980).

G6PD 'CLICHY'. See Boivin and Galand (1968).

G6PD CLINIC. In a young patient with chronic nonspherocytic hemolytic anemia and familial amyloidotic polyneuropathy, Vives-Corrons et al. (1989) identified a new variant with a markedly acidic pH optimum. It bore some similarity in its molecular characteristics to G6PD Bangkok and G6PD Duarte.

G6PD COLOMIERS. See Vergnes et al. (1981).

G6PD COLUMBUS. See Pinto et al. (1966).

G6PD CORINTH. Yoshida, A.: unpublished, 1975.

G6PD CORNELL. See Miller and Wollman (1974) and Fairbanks et al. (1980). Fairbanks et al. (1980) demonstrated that G6PD Chicago and G6PD Cornell are the same variant; they had been described previously in different members of a single large kindred.

G6PD CUIABA. In a 33-year-old male of Portuguese extraction who developed hemolytic anemia after acetaminophen and acetylsalicylic acid ingestion, Barretto and Nonoyama (1987) found a variant G6PD which had normal activity and normal electrophoretic mobility, but unusually high K(m) for glucose-6-phosphate, high K(i) for NADPH, and decreased thermal stability.

G6PD 'DAKAR'. See Kahn et al. (1971, 1973).

G6PD DALLAS. Beutler, E.; Frenkel, E. P.; Forman, L.: unpublished, 1987.

G6PD DEBROUSSE (G6PD CONSTANTINE, FORMERLY). See Kissin and Cotte (1970) and Sansone et al. (1975).

G6PD DJYNET. See Krasnopolskaya and Bochkov (1982).

G6PD DOTHAN. See Prchal et al. (1979).

G6PD DUARTE. See Beutler et al. (1968).

G6PD DUBLIN. See McCann et al. (1980).

G6PD DUSHANBA I. See Krasnopolskaya and Bochkov (1982).

G6PD DUSHANBA II. See Krasnopolskaya and Bochkov (1982).

G6PD DUSHANBA III. See Krasnopolskaya and Bochkov (1982).

G6PD EAST AFRICAN. See Othieno-Obel (1972).

G6PD EAST HARLEM. See Feldman et al. (1977).

G6PD 'EKITI'. See Usanga et al. (1977).

G6PD EL-FAYOUM. See McCurdy et al. (1974).

G6PD EL-KHARGA. See McCurdy et al. (1974).

G6PD EL MORRO. See McCurdy et al. (1973).

G6PD ENGLEWOOD. See Rattazzi et al. (1971).

G6PD 'ENSLEY'. See Nsouly and Prchal (1981).

G6PD 'ESPOO'. See Vuopio et al. (1975).

G6PD FERRANDINA. See Calabro et al. (1990).

G6PD FERRARA. See Carandina et al. (1976).

G6PD FERRARA II. See De Flora et al. (1981) and Sansone et al. (1981).

G6PD 'FERRARA III'. See Perroni et al. (1982).

G6PD FORT PIERCE. Phyliky, R. L.; Nishimura, R. A. and Beutler, E.: unpublished, 1983.

G6PD FORT WORTH. See Mills et al. (1975).

G6PD 'FRANKFURT'. Nowicki et al. (1974).

G6PD FREIBURG. See Weinreich et al. (1968) and Busch and Boie (1970).

G6PD FUKUOKA. This variant was found in a 77-year-old male with drug-induced hemolysis (Fujii et al., 1984). Enzyme activity was 6.4% of normal and the patient's G6PD had abnormal electrophoretic mobility and thermal instability.

G6PD FUKUSHIMA. Miwa et al. (1978) described this 'deficiency' mutant, which leads to chronic hemolytic anemia. It was slow-moving electrophoretically, like G6PD Kurume, from which it differed by low utilization of deamino-NADP and normal pH curve. The proband, a 33-year-old male, had 2.8% of normal enzyme activity and mild hemolytic anemia. Miwa et al. (1978) stated that 46 variants had previously been classified as class 1, with severe enzyme deficiency leading to chronic nonspherocytic hemolytic anemia.

G6PD GABROVIZZA. See Ventura et al. (1984).

G6PD 'GALLIERA'. See Perroni et al. (1982).

G6PD GALLURA. See Sansone et al. (1975).

G6PD 'GALVESTON'. See Alperin and Mills (1972).

G6PD 'GAMBIA'. Welch et al. (1978) found a gene frequency of 0.024 among 1,109 persons examined in The Gambia. This is a slow electrophoretic variant with reduced enzyme activity.

G6PD GAOMIN. See Du et al. (1988).

G6PD GAOZHOU. See Du et al. (1988).

G6PD GENOVA. See Gaetani et al. (1990).

G6PD GOODENOUGH. See Chockkalingam et al. (1982).

G6PD GOTZE DELCHEV. See Shatskaya et al. (1980). G6PD GRAND PRAIRIE. See Cederbaum and Beutler (1975).

G6PD GREAT LAKES. Beutler, E. and Maurer, H. S.: unpublished, 1984.

G6PD GUANGZHOU. See Du et al. (1988).

G6PD GUANTANAMO. See Gutierrez et al. (1987). G6PD 'GUIBA'. See Weimer et al. (1981).

G6PD HAAD YAI. See Panich and Na-Nakorn (1980).

G6PD 'HAMBURG'. See Gahr and Schroter (1974).

G6PD HAMM. See Gahr et al. (1976).

G6PD 'HANOI'. See Toncheva (1986).

G6PD HAWAII. Beutler, E. and Matsumoto, F.: unpublished, 1975.

G6PD HAYEM. See Kahn et al. (1974). G6PD HEIAN. See Nakai and Yoshida (1974).

G6PD HEKTOEN. Substitution of tyrosine for histidine (Dern et al., 1969).

G6PD HELSINKI. See Vuopio et al. (1973) and Harkonen and Vuopio (1974). Cohn et al. (1979) described severe hemolytic anemia in 2 Danish boys, who showed deficiency of G6PD. The enzyme had characteristics possibly identical to those of G6PD Helsinki.

G6PD HILLBROW. See Cayanis et al. (1975).

G6PD 'HIROSHIMA-1'. See Kageoka et al. (1985).

G6PD 'HIROSHIMA-2'. See Kageoka et al. (1985).

G6PD 'HIROSHIMA-3'. See Kageoka et al. (1985).

G6PD HOFU. See Miwa et al. (1977).

G6PD HONG KONG. See Wong et al. (1965) and Chan et al. (1972).

G6PD HONG KONG POKFULAM. See Chan et al. (1972).

G6PD HOTEL DIEU. See Kahn et al. (1977).

G6PD HUALIEN. McCurdy, P. R.: unpublished, 1975.

G6PD HUALIEN-CHI. McCurdy, P. R.: unpublished, 1975.

G6PD HUAZHOU. See Du et al. (1988).

G6PD HUIYANG. See Du et al. (1988).

G6PD HUNTSVILLE. See Hall et al. (1988).

G6PD HURON. See Ravindranath and Beutler (1987).

G6PD IBADAN-AUSTIN. See Long et al. (1965).

G6PD IJEBU-ODE. See Luzzatto and Afolayan (1968).

G6PD INDIANAPOLIS. Beutler, E.; Forman, L.; Gelbart, T.: unpublished, 1985.

G6PD INDONESIA. See Kirkman and Eng (1969).

G6PD INHAMBANE. See Reys et al. (1970).

G6PD INTANON. See Panich (1974).

G6PD ISERLOHN. Unstable enzyme. See Eber et al. (1985).

G6PD ITA-BALE. See Long et al. (1965).

G6PD IWATE. See Kanno et al. (1987).

G6PD JACKSON. See Thigpen et al. (1974).

G6PD JALISCO. See Vaca et al. (1985).

G6PD JOHANNESBURG. See Balinsky et al. (1973).

G6PD 'JUNUT'. See Shatskaya et al. (1980).

G6PD KABYLE. See Kaplan et al. (1967).

G6PD KALUAN. See Chockkalingam and Board (1980).

G6PD KALUGA. See Shatskaya et al. (1976).

G6PD KAMIUBE. See Nakatsuji and Miwa (1979).

G6PD KAN. See Panich (1973).

G6PD KANAZAWA. This variant, found by Kitao et al. (1982) in a Japanese male with chronic nonspherocytic hemolytic anemia, has normal electrophoretic mobility, normal Km for glucose-6-phosphate and NADP, and normal utilization of the substrate 2-deoxyglucose-6-phosphate and deamino-NADP. It shows decreased thermal stability and a biphasic pH curve. G6PD KAR KAR. See Chockkalingam et al. (1982).

G6PD KARDISTA. Stamatoyannopoulos, G.: unpublished, 1975.

G6PD KEPHALONIA. See Rattazzi et al. (1969).

G6PD KEROVOGRAD. See Krasnopolskaya and Bochkov (1982). G6PD 'KHARTOUM'. See Samuel et al. (1981).

G6PD 'KILGORE'. See Alperin and Mills (1972).

G6PD KING COUNTY. Yoshida, A.: unpublished, 1975.

G6PD KIROVOGRAD. See Shatskaya et al. (1976).

G6PD KIWA. See Nakatsuji and Miwa (1979).

G6PD KOBE. See Fujii et al. (1981).

G6PD KONAN. See Nakatsuji and Miwa (1979).

G6PD KREMENCHUNG. See Cherniak et al. (1977) and Tokarev et al. (1978).

G6PD KUANYAMA. See Balinsky et al. (1974).

G6PD KURUME. A 'deficiency' mutation, this variant leads to chronic hemolytic anemia. It is electrophoretically slow-moving. The proband was a 17-year-old male whose red cells had only 0.8% normal enzyme activity (Miwa et al., 1978). The enzyme showed normal KmG6P, normal KmNADP, low KiNADP, normal utilization of 2-deoxy-G6P and deamino-NADP, very low heat stability, and a biphasic pH curve.

G6PD 'KYOTO'. See Kojima (1972). G6PD LAGHOUAT. See Benabadji et al. (1978).

G6PD LAGUNA. Although the proband was anemic, the absence of anemia in relatives with the same G6PD variant suggested that the association was coincidental (Weimer et al., 1984). The characteristics of the mutant enzyme, including slower electrophoretic mobility, were described.

G6PD 'LANLATE'. See Usanga et al. (1977).

G6PD LAOS. Smith, J. W. and Beutler, E.: unpublished, 1981.

G6PD LAWNDALE. See Grossman et al. (1966).

G6PD LEVADIA. See Stamatoyannopoulos et al. (1970).

G6PD LIFTA. See Ramot et al. (1969).

G6PD LINCOLN PARK. See Honig et al. (1979).

G6PD LINDA VISTA. Smith, J. W. and Beutler, E.: unpublished, 1981.

G6PD 'LIZU-BAISHA'. See Du (1981).

G6PD LONG PRAIRIE. See Johnson et al. (1977).

G6PD LONG XUYEN. See Panich et al. (1980).

G6PD LOS ANGELES. See Beutler and Matsumoto (1977).

G6PD LOURENZO MARQUES. See Reys et al. (1970). G6PD LOZERE. See Vergnes et al. (1976).

G6PD LUBLIN. See Pawlak et al. (1970).

G6PD LUZ-SAINT SAUVEUR. See Vergnes et al. (1973).

G6PD LYNN (G6PD YUGOSLAVIA, FORMERLY). Beutler, E. and Lind, S.: unpublished, 1987.

G6PD MADANG. See Chockkalingam et al. (1982).

G6PD MADISON. See Shows et al. (1964).

G6PD MADRONA. See Hook et al. (1968).

G6PD MAINOKI. See Chockkalingam et al. (1982).

G6PD 'MALI'. See Kahn et al. (1971).

G6PD MAMMOLA. See Perroni et al. (1982).

G6PD MANCHESTER. See Milner et al. (1974).

G6PD MANDANG. See Chockkalingam et al. (1982).

G6PD MANJACAZE. See Reys et al. (1970).

G6PD MANUS. See Chockkalingam et al. (1982).

G6PD MARKHAM. See Kirkman et al. (1968).

G6PD 'MARTINIQUE'. See Kahn et al. (1971).

G6PD MARTINIQUE-LIKE. See Krasnopolskaya et al. (1977).

G6PD MATAM. See Kahn et al. (1975).

G6PD MELISSA. Stamatoyannopoulos, G.: unpublished, 1975.

G6PD MENORCA. See Vives-Corrons and Pujades (1982).

G6PD MERCURY. Beutler, E. and Taylor, G. P.: unpublished, 1982.

G6PD MEXICO. See Lisker et al. (1972).

G6PD MIAOZU-BAISHA. See Du et al. (1984).

G6PD MILWAUKEE. See Westring and Pisciotta (1966).

G6PD MINAS GERAIS. See Azevedo and Yoshida (1969).

G6PD MINNEAPOLIS. Johnson, G. J. and Beutler, E.: unpublished, 1980.

G6PD 'MISENO'. See Colonna-Romano et al. (1985).

G6PD MISSOULA. See Wilson (1976).

G6PD MOOSBURG. See Pekrun et al. (1989).

G6PD MORELIA. Class 4. First in class with a high Km for NADP and a low Ki for NADPH. See Vaca et al. (1985).

G6PD MOSCOW. See Batischev et al. (1977).

G6PD MURET. See Vergnes et al. (1981).

G6PD MUSASHINO. See Kumakawa et al. (1987).

G6PD NAGANO. This variant is associated with infection-induced hemolysis and chronic hemolytic anemia due to markedly impaired enzyme activity and thermal instability (Takahashi et al., 1982).

G6PD 'NAGASAKI-1'. See Kageoka et al. (1985).

G6PD 'NAGASAKI-2'. See Kageoka et al. (1985).

G6PD 'NAGASAKI-3'. See Kageoka et al. (1985).

G6PD 'NANCY'. See Streiff and Vigneron (1971).

G6PD NANHAI. See Du et al. (1988).

G6PD NAPOLI. See De Flora et al. (1981).

G6PD NEDELINO. See Toncheva and Tzoneva (1984).

G6PD NEW GUINEA-II. See Rattazzi et al. (1971).

G6PD NEW YORK. See Rattazzi et al. (1971).

G6PD N-PATHOM. See Panich (1974) and Panich and Na-Nakorn (1980).

G6PD N-SAWAN. See Panich and Na-Nakorn (1980).

G6PD NUCUS. See Yermakov et al. (1981).

G6PD NUHA. See Krasnopolskaya and Bochkov (1982).

G6PD 'NUKHA'. See Shatskaya et al. (1980).

G6PD OGIKUBO. See Miwa et al. (1978).

G6PD OGORI. See Lisker et al. (1977). G6PD OHIO. See Pinto et al. (1966).

G6PD OKHUT I. See Krasnopolskaya et al. (1977).

G6PD OKHUT II. See Krasnopolskaya et al. (1977).

G6PD OKLAHOMA. See Kirkman and Riley (1961) and Nance (1964).

G6PD ONODA. Nakashima, K.: unpublished, 1978.

G6PD ORCHOMENOS. See Stamatoyannopoulos et al. (1971).

G6PD PADREW. See Panich and Na-Nakorn (1980).

G6PD PALAKAU. See Chockkalingam et al. (1982).

G6PD 'PALEPOLI'. See Colonna-Romano et al. (1985).

G6PD 'PALLONETTO'. See Colonna-Romano et al. (1985).

G6PD 'PALMI I'. See Perroni et al. (1982).

G6PD 'PALMI II'. See Perroni et al. (1982).

G6PD PANAMA. See Beutler et al. (1974).

G6PD PANAY. See Fernandez and Fairbanks (1968).

G6PD PANAY-LIKE.

G6PD 'PARIS'. See Boivin and Galand (1968).

G6PD PEA RIDGE. See Fairbanks et al. (1980).

G6PD 'PETILIA'. See Sansone et al. (1981) and Perroni et al. (1982).

G6PD PETRICH. See Shatskaya et al. (1980).

G6PD PINAR DEL RIO. See Gonzalez et al. (1977).

G6PD PISTICCI. See Viglietto et al. (1990) and Calabro et al. (1990).

G6PD POMPTON PLAINS. Beutler, E.; Davis, S.; Forman, L. and Gelbart, T.: unpublished, 1985.

G6PD POPONDETTA. See Chockkalingam et al. (1982).

G6PD PORBANDAR. See Cayanis et al. (1977).

G6PD 'PORDENONE'. See Sansone et al. (1981) and Perroni et al. (1982).

G6PD PORT ELIZABETH. See Balinsky et al. (1973).

G6PD PORT-ROYAL. See Kaplan et al. (1971).

G6PD PORTO ALEGRE. See Hutz et al. (1977).

G6PD 'POSILIPPO'. See Colonna-Romano et al. (1985).

G6PD POZNAN. See Pawlak et al. (1975).

G6PD 'POZZALLO'. See Perroni et al. (1982).

G6PD PUERTO RICO. See McCurdy et al. (1973).

G6PD QING-BAILJIANG. See Du et al. (1988).

G6PD RAMAT-GAN. See Ramot et al. (1969).

G6PD REGAR. See Ermakov et al. (1983).

G6PD REGENSBURG. See Eber et al. (1985).

G6PD 'RENNES'. See Picat et al. (1980).

G6PD ROTTERDAM. See Rattazzi et al. (1971).

G6PD RUDOSEM. See Toncheva and Tzoneva (1984).

G6PD RUSSIAN-MOSCOW. See Krasnopolskaya and Bochkov (1982).

G6PD SALATA. See Chockkalingam and Board (1980).

G6PD SAMANDAG. See Aksoy et al. (1987).

G6PD SAN DIEGO. See Howell et al. (1972).

G6PD SAN FRANCISCO. See Mentzer et al. (1980).

G6PD SAN JOSE. See Castro and Snyder (1974).

G6PD SAN JUAN. See McCurdy et al. (1973).

G6PD SANTA BARBARA. Kidder, W. R. and Beutler, E.: unpublished, 1979.

G6PD SAPPORO. See Fujii et al. (1981).

G6PD 'SCHWABEN'. See Benohr et al. (1971).

G6PD 'S.DONA'. See Perroni et al. (1982).

G6PD SEATTLE. See Kirkman et al. (1965).

G6PD SELIM. See Shatskaya et al. (1975).

G6PD SENDAGI. This variant was associated with chronic nonspherocytic hemolytic anemia in a 2-year-old Japanese male in whom upper respiratory infection precipitated a hemolytic crisis (Morisaki et al.,1983).

G6PD SHEKII. See Krasnopolskaya et al. (1977).

G6PD SHIRIN-BULAKH. See Krasnopolskaya et al. (1977).

G6PD SIRIRAJ. See Panich et al. (1972).

G6PD SIWA. See McCurdy et al. (1974).

G6PD SONGKHLA. See Panich and Na-Nakorn (1980).

G6PD S-SAKORN. See Panich (1980).

G6PD ST. LOUIS. See Kahn et al. (1974).

G6PD STEILACOM. Yoshida, A.; Baur, E. and Voigtlander, B.: unpublished, 1975.

G6PD 'STELLA'. See Colonna-Romano et al. (1985).

G6PD 'STRASBOURG'. See Waitz et al. (1970).

G6PD SWIT. See Chockkalingam et al. (1982).

G6PD TACOMA. Yoshida, A. and Baur, E.: unpublished, 1975.

G6PD TACOMA-LIKE. See Vergnes et al. (1975).

G6PD TAHTA. See McCurdy et al. (1974).

G6PD TAIPEI-HAKKA. See McCurdy et al. (1970).

G6PD 'TAIWAN-AMI 5'. See McCurdy et al. (1970).

G6PD 'TAIWAN-AMI 6'. See McCurdy et al. (1970).

G6PD TARSUS. See Gahr et al. (1976).

G6PD TASHKENT. See Yermakov et al. (1981).

G6PD TEHERAN. McCurdy, P. R.: unpublished, 1965.

G6PD TEL HASHOMER. See Ramot and Brok (1964) and Kirkman et al. (1969).

G6PD TENGANAN. See Chockkalingam et al. (1982).

G6PD THENIA. See Benabadji et al. (1978).

G6PD THESSALONIKI. Koliakos et al. (1989) found a new variant in a 70-year-old patient with idiopathic myelofibrosis. This disorder, formerly called agnogenic myeloid metaplasia, is a myeloproliferative disease with clonal origin in a malignant pluripotent stem cell. Bone marrow fibrosis is a secondary process. The patient was thought to be heterozygous since her only son had normal G6PD. That she showed severe G6PD deficiency was taken to indicate that the normal X chromosome was active in the original cell that underwent malignancy.

G6PD THESSALY. See Stamatoyannopoulos et al. (1970).

G6PD TITTERI. See Benabadji et al. (1978).

G6PD TITUSVILLE. Csepreghy et al. (1989) described a new G6PD variant in a 7-month-old black male and his mother. The proband had had a transient hemolytic episode.

G6PD TOKUSHIMA. See Miwa et al. (1976).

G6PD TOKYO. See Miwa et al. (1976).

G6PD TORONTO. See Crookston et al. (1973).

G6PD TORRANCE. See Tanaka and Beutler (1969).

G6PD TOULOUSE. See Vergnes et al. (1974).

G6PD 'TRAPANI'. See Sansone et al. (1981) and Perroni et al. (1982).

G6PD TRINACRIA. See Sansone et al. (1977).

G6PD TRIPLER. See Engstrom and Beutler (1970).

G6PD TSUKUI. See Ogura et al. (1988).

G6PD TUBINGEN. See Benohr and Waller (1970).

G6PD TURSI. See Viglietto et al. (1990) and Calabro et al. (1990).

G6PD UBE. See Nakashima et al. (1977).

G6PD UNION. See Yoshida et al. (1970).

G6PD 'UNION-MARKHAM'. See Stamatoyannopoulos et al. (1971).

G6PD 'UNNAMED'. See Othieno-Obel (1972).

G6PD 'VARADERO'. See Estrada et al. (1982).

G6PD VELLETRI. See Mandelli et al. (1977).

G6PD VIENTIANE. See Kahn et al. (1978).

G6PD 'VIN FU'. See Toncheva (1986).

G6PD WAKAYAMA. This variant was found in a 16-month-old boy with 4.5% of normal enzyme activity and mild hemolytic anemia (Miwa et al., 1978). Electrophoretically, it is slow-moving like G6PD Kurume, from which it differs by a normal pH curve. In addition to the 4 slow variants reported by Miwa et al. (1978), 5 had previously been reported: Alhambra, Atlanta, Hong Kong Pokfulam, Manchester, and Tokyo. G6PD WASHINGTON. McCurdy, P. R.: unpublished, 1975.

G6PD WATERLOO. Beutler, E. and Phyliky, R. L.: unpublished, 1978.

G6PD WAYNE. See Ravindranath and Beutler (1987).

G6PD WEST BENGAL. See Azevedo et al. (1968).

G6PD WEST TOWN. This variant causes chronic nonspherocytic anemia which is compensated except following infections or exposure to an oxidant drug (Honig et al., 1979).

G6PD WESTERN. Yoshida, A. and Baur, E.: unpublished, 1975.

G6PD WEWAK. See Chockkalingam et al. (1982).

G6PD WORCESTER. Snyder et al. (1970) described a family in which a new variant form of G6PD was associated with congenital nonspherocytic hemolytic anemia and optic atrophy in 3 males related as first cousins once removed. Blindness developed rapidly in the teens.

G6PD 'WROCLAW'. See Kwiatkowska and Kacprzak-Bergman (1971).

G6PD YAMAGUCHI. This variant was found in an 8-year-old boy who had 3.5% of normal enzyme activity and moderate hemolytic anemia (Miwa et al., 1978). Electrophoretically, it is slow-moving, like G6PD Kurume, from which it differs by high Km NADP, high deamino-NADP utilization, and an abnormal pH curve of a different type (with narrow peak at pH 8.76).

G6PD YANGORU. See Chockkalingam et al. (1982).

G6PD YOKOHAMA. See Miwa et al. (1978).

G6PD 'ZAEHRINGEN'. See Witt and Yoshioka (1972).

G6PD ZAKATALY. See Krasnopolskaya et al. (1977).

G6PD ZHITOMIR. See Shatskaya et al. (1976).


See Also:

REFERENCES

  1. Adam, A., Tippett, P., Gavin, J., Noades, J., Sanger, R., Race, R. R. The linkage relation of Xg to G6PD in Israelis: the evidence of a second series of families. Ann. Hum. Genet. 30: 211-218, 1967. [PubMed: 6032005, related citations] [Full Text]

  2. Ahluwalia, A., Corcoran, C. M., Vulliamy, T. J., Ishwad, C. S., Naidu, J. M., Argusti, A., Stevens, D. J., Mason, P. J., Luzzatto, L. G6PD Kalyan and G6PD Kerala; two deficient variants in India caused by the same 317 glu-to-lys mutation. Hum. Molec. Genet. 1: 209-210, 1992. [PubMed: 1303182, related citations] [Full Text]

  3. Aksoy, K., Yuregir, G. T., Dikmen, N., Unlukurt, I. Three new G6PD variants, G6PD Adana, G6PD Samandag, and G6PD Balcali in Cukurova, Turkey. Hum. Genet. 76: 199-201, 1987. [PubMed: 3610149, related citations] [Full Text]

  4. Alfinito, F., Cimmino, A., Ferraro, F., Cubellis, M. V., Vitagliano, L., Francese, M., Zagari, A., Rotoli, B., Filosa, S., Martini, G. Molecular characterization of G6PD deficiency in Southern Italy: heterogeneity, correlation genotype-phenotype and description of a new variant (G6PD Neapolis). Brit. J. Haemat. 98: 41-46, 1997. [PubMed: 9233561, related citations] [Full Text]

  5. Alperin, J. B., Mills, G. C. New variants of glucose-6-phosphate dehydrogenase (G6PD). (Abstract) Clin. Res. 20: 76, 1972.

  6. Azevedo, E., Kirkman, H. N., Morrow, A. C., Motulsky, A. G. Variants of red cell glucose-6-phosphate dehydrogenase among Asiatic Indians. Ann. Hum. Genet. 31: 373-379, 1968. [PubMed: 5673160, related citations] [Full Text]

  7. Azevedo, E. S., Yoshida, A. Brazilian variant of glucose-6-phosphate dehydrogenase (GD Minas Gerais). Nature 222: 380-382, 1969. [PubMed: 5782116, related citations] [Full Text]

  8. Babalola, A. O. G., Beetlestone, J. G., Luzzatto, L. Genetic variants of human erythrocyte glucose-6-phosphate dehydrogenase: kinetic and thermodynamic parameters of variants A, B, and A- in relation to quaternary structure. J. Biol. Chem. 251: 2993-3002, 1976. [PubMed: 5448, related citations]

  9. Balinsky, D., Cayanis, E., Carter, G., Jenkins, T., Bersohn, I. A new variant of human erythrocyte glucose-6-phosphate dehydrogenase: G6PD Port Elizabeth. Int. J. Biochem. 4: 235-244, 1973.

  10. Balinsky, D., Gomperts, E., Cayanis, E., Jenkins, T., Bryer, D., Bersohn, I., Metz, J. Glucose 6-phosphate dehydrogenase Johannesburg: a new variant with reduced activity in a patient with congenital non-spherocytic haemolytic anaemia. Brit. J. Haemat. 25: 385-392, 1973. [PubMed: 4147554, related citations] [Full Text]

  11. Balinsky, D., Rootman, A. J., Nurse, G. T., Cayanis, E., Lane, A., Jenkins, T., Bersohn, I. G6PD Kuanyama: a new variant of human erythrocyte glucose 6-phosphate dehydrogenase showing slower than normal electrophoretic mobility. S. Afr. J. Med. Sci. 39: 5-13, 1974. [PubMed: 4153368, related citations]

  12. Barisic, M., Korac, J., Pavlinac, I., Krzelj, V., Marusic, E., Vulliamy, T., Terzic, J. Characterization of G6PD deficiency in southern Croatia: description of a new variant, G6PD Split. J. Hum. Genet. 50: 547-549, 2005. [PubMed: 16143877, related citations] [Full Text]

  13. Barretto, O. C., Nonoyama, K. Gd(+) Cuiaba, a new rare glucose-6-phosphate dehydrogenase variant presenting normal activity. Hum. Genet. 77: 201-202, 1987. [PubMed: 3653894, related citations] [Full Text]

  14. Batischev, A. I., Chernyak, N. B., Torakev, Y. N. Detection of a new abnormal variant of glucose-6-phosphate dehydrogenase in human red cells. Biull. Eksp. Biol. Med. 84: 728-731, 1977. [PubMed: 23188, related citations]

  15. Ben-Bassat, J., Ben-Ishay, D. Hereditary hemolytic anemia associated with glucose-6-phosphate dehydrogenase deficiency (Mediterranean type). Isr. J. Med. Sci. 5: 1053-1059, 1969. [PubMed: 5369703, related citations]

  16. Benabadji, M., Merad, F., Benmoussa, M., Trabuchet, G., Junien, C., Dreyfus, J. C., Kaplan, J. C. Heterogeneity of glucose-6-phosphate dehydrogenase deficiency in Algeria. Hum. Genet. 40: 177-184, 1978. [PubMed: 624546, related citations] [Full Text]

  17. Benohr, H. C., Klumpp, F., Waller, H. D. Glucose-6-phosphat-Dehydrogenase Typ Schwaben. Dtsch. Med. Wschr. 96: 1029-1032, 1971. [PubMed: 5580403, related citations] [Full Text]

  18. Benohr, H. C., Waller, H. D., Arnold, H., Blume, K. G., Lohr, G. W. Glucose-6-P-Dehydrogenase Typ Bodensee (eine neue Enzymvariante). Klin. Wschr. 49: 1058-1062, 1971. [PubMed: 5115324, related citations] [Full Text]

  19. Benohr, H. C., Waller, H. D. Eigenschaften der Glucose-6-p-dehydrogenase, Typ Tubingen. Klin. Wschr. 48: 71-74, 1970. [PubMed: 4398790, related citations] [Full Text]

  20. Beutler, E., Grooms, A. M., Morgan, S. K., Trinidad, F. Chronic severe hemolytic anemia due to G-6-PD Charleston: a new deficiency variant. J. Pediat. 80: 1005-1009, 1972. [PubMed: 4402065, related citations] [Full Text]

  21. Beutler, E., Hartman, K., Gelbart, T., Forman, L. G-6-PD Walter Reed: possible insight into 'structural' NADP in G-6-PD. Am. J. Hemat. 23: 25-30, 1986. [PubMed: 3740052, related citations] [Full Text]

  22. Beutler, E., Keller, J. W., Matsumoto, F. A new glucose 6-phosphate dehydrogenase (G6PD) variant associated with nonspherocytic hemolytic anemia: G6PD Atlanta. I.R.C.S. 4: 479, 1976.

  23. Beutler, E., Kuhl, W., Gelbart, T., Forman, L. DNA sequence abnormalities of human glucose-6-phosphate dehydrogenase variants. J. Biol. Chem. 266: 4145-4150, 1991. [PubMed: 1999409, related citations]

  24. Beutler, E., Kuhl, W., Ramirez, E., Lisker, R. Some Mexican glucose-6-phosphate dehydrogenase variants revisited. Hum. Genet. 86: 371-374, 1991. [PubMed: 1999339, related citations] [Full Text]

  25. Beutler, E., Kuhl, W., Saenz, G. F., Rodriguez, W. Mutation analysis of glucose-6-phosphate dehydrogenase (G6PD) variants in Costa Rica. Hum. Genet. 87: 462-464, 1991. [PubMed: 1879833, related citations] [Full Text]

  26. Beutler, E., Kuhl, W., Vives-Corrons, J.-L., Prchal, J. T. Molecular heterogeneity of glucose-6-phosphate dehydrogenase A-. Blood 74: 2550-2555, 1989. [PubMed: 2572288, related citations]

  27. Beutler, E., Kuhl, W. Linkage between a PvuII restriction fragment length polymorphism and G6PD A- (202A/376G): evidence for a single origin of the common G6PD A- mutation. Hum. Genet. 85: 9-11, 1990. [PubMed: 1972698, related citations] [Full Text]

  28. Beutler, E., Kuhl, W. The NT 1311 polymorphism of G6PD: G6PD Mediterranean mutation may have originated independently in Europe and Asia. Am. J. Hum. Genet. 47: 1008-1012, 1990. [PubMed: 1978554, related citations]

  29. Beutler, E., Mathai, C. K., Smith, J. E. Biochemical variants of glucose-6-phosphate dehydrogenase giving rise to congenital nonspherocytic hemolytic disease. Blood 31: 131-150, 1968. [PubMed: 5643703, related citations]

  30. Beutler, E., Matsumoto, F., Daiber, A. Nonspherocytic hemolytic anemia due to G-6-PD Panama. I.R.C.S. 2: 1389, 1974.

  31. Beutler, E., Matsumoto, F. A new glucose 6-phosphate dehydrogenase variant: G6PD (-) Los Angeles. I.R.C.S. 5: 89, 1977.

  32. Beutler, E., Rosen, R. Nonspherocytic congenital hemolytic anemia due to a new G-6-PD variant: G-6-PD Alhambra. Pediatrics 45: 230-235, 1970. [PubMed: 5413384, related citations]

  33. Beutler, E., Westwood, B., Prchal, J. T., Vaca, G., Bartsocas, C. S., Baronciani, L. New glucose-6-phosphate dehydrogenase mutations from various ethnic groups. Blood 80: 255-256, 1992. [PubMed: 1611091, related citations]

  34. Beutler, E., Yoshida, A. Human glucose-6-phosphate dehydrogenase variants: a supplementary tabulation. Ann. Hum. Genet. 37: 151-156, 1973. [PubMed: 4768554, related citations] [Full Text]

  35. Beutler, E. Glucose 6-phosphate dehydrogenase deficiency, a new Indian variant, G6PD Jammu. In: Sen, N. N.; Basu, A. K. (eds.): Trends in Haematology. Calcutta: Chatterjea Memorial Committee (pub.) 1975. Pp. 279-283.

  36. Beutler, E. Selectivity of proteases as a basis for tissue distribution of enzymes in hereditary deficiencies. Proc. Nat. Acad. Sci. 80: 3767-3768, 1983. [PubMed: 6344088, related citations] [Full Text]

  37. Beutler, E. Glucose-6-phosphate dehydrogenase deficiency. New Eng. J. Med. 324: 169-174, 1991. [PubMed: 1984194, related citations] [Full Text]

  38. Boivin, P., Galand, C. Nouvelles variantes de la glucose-6-phosphate dehydrogenase erythrocytaire. Rev. Franc. Etud. Clin. Biol. 13: 30-39, 1968. [PubMed: 5653554, related citations]

  39. Botha, M. C., Dern, R. J., Mitchell, M., West, C., Beutler, E. G6PD Capetown, a variant of glucose-6-phosphate dehydrogenase. Am. J. Hum. Genet. 21: 547-551, 1969. [PubMed: 5365756, related citations]

  40. Boyer, S. H., Graham, J. B. Linkage between the X chromosome loci for glucose-6-phosphate dehydrogenase electrophoretic variation and hemophilia A. Am. J. Hum. Genet. 17: 320-324, 1965. [PubMed: 17948498, related citations]

  41. Boyer, S. H., Porter, I. H., Weilbaecher, R. G. Electrophoretic heterogeneity of glucose-6-phosphate dehydrogenase and its relationship to enzyme deficiency in man. Proc. Nat. Acad. Sci. 48: 1868-1876, 1962. [PubMed: 14014720, related citations] [Full Text]

  42. Busch, D., Boie, K. Glucose-6-phosphate-dehydrogenase-Defect in Deutschland. II. Bigenschabten des Enzyms (Typ Freiburg). Klin. Wschr. 48: 74-78, 1970. [PubMed: 4398791, related citations] [Full Text]

  43. Calabro, V., Giacobbe, A., Vallone, D., Montanaro, V., Cascone, A., Filosa, S., Battistuzzi, G. Genetic heterogeneity at the glucose-6-phosphate dehydrogenase locus in southern Italy: a study on a population from the Matera district. Hum. Genet. 86: 49-53, 1990. [PubMed: 2253938, related citations] [Full Text]

  44. Calabro, V., Mason, P. J., Filosa, S., Civitelli, D., Cittadella, R., Tagarelli, A., Martini, G., Brancati, C., Luzzatto, L. Genetic heterogeneity of glucose-6-phosphate dehydrogenase deficiency revealed by single-strand conformation and sequence analysis. Am. J. Hum. Genet. 52: 527-536, 1993. [PubMed: 8447319, related citations]

  45. Cappadoro, M., Giribaldi, G., O'Brien, E., Turrini, F., Mannu, F., Ulliers, D., Simula, G., Luzzatto, L., Arese, P. Early phagocytosis of glucose-6-phosphate dehydrogenase (G6PD)-deficient erythrocytes parasitized by Plasmodium falciparum may explain malaria protection in G6PD deficiency. Blood 92: 2527-2534, 1998. [PubMed: 9746794, related citations]

  46. Cappellini, M. D., Fiorelli, G. Glucose-6-phosphate dehydrogenase deficiency. Lancet 371: 64-74, 2008. [PubMed: 18177777, related citations] [Full Text]

  47. Cappellini, M. D., Sampietro, M., Toniolo, D., Carandina, G., Pittalis, S., Martinez di Montemuros, F., Tavazzi, D., Fiorelli, G. Biochemical and molecular characterization of a new sporadic glucose-6-phosphate dehydrogenase variant described in Italy: G6PD Modena. Brit. J. Haemat. 87: 209-211, 1994. [PubMed: 7947250, related citations] [Full Text]

  48. Carandina, G., Moretto, E., Zecchi, G., Conighi, C. Glucose 6-phosphate dehydrogenase Ferrara. A new variant of G6PD identified in Northern Italy. Acta Haemat. 56: 116-122, 1976. [PubMed: 823756, related citations] [Full Text]

  49. Castro, A. M., Snyder, L. M. G6PD San Jose: a new variant characterized by NADPH inhibition studies. Humangenetik 21: 361-363, 1974. [PubMed: 4151903, related citations] [Full Text]

  50. Cayanis, E., Gomperts, E. D., Balinsky, D., Disler, P., Myers, A. G6PD Hillbrow: a new variant of glucose-6-phosphate dehydrogenase associated with drug-induced haemolytic anaemia. Brit. J. Haemat. 30: 343-350, 1975. [PubMed: 1201217, related citations] [Full Text]

  51. Cayanis, E., Lane, A. B., Jenkins, T., Nurse, G. T., Balinsky, D. Glucose-6-phosphate dehydrogenase Porbandar: a new slow variant with slightly reduced activity in a South African family of Indian descent. Biochem. Genet. 15: 765-773, 1977. [PubMed: 921743, related citations] [Full Text]

  52. Cazzola, M., Bergamaschi, G. X-linked Wiskott-Aldrich syndrome in a girl. (Letter) New Eng. J. Med. 338: 1850 only, 1998. [PubMed: 9634367, related citations] [Full Text]

  53. Cederbaum, A. I., Beutler, E. Nonspherocytic hemolytic anemia due to G6PD Grand Prairie. I.R.C.S. 3: 579, 1975.

  54. Chalvam, R., Kedar, P. S., Colah, R. B., Ghosh, K., Mukherjee, M. B. A novel R198H mutation in the glucose-6-phosphate dehydrogenase gene in the tribal groups of the Nilgiris in southern India. J. Hum. Genet. 53: 181-184, 2008. [PubMed: 18043863, related citations] [Full Text]

  55. Chalvam, R., Mukherjee, M. B., Colah, R. B., Mohanty, D., Ghosh, K. G6PD Namoru (208T-C) is the major polymorphic variant in the tribal populations in southern India. (Letter) Brit. J. Haemat. 136: 512-513, 2007. [PubMed: 17233850, related citations] [Full Text]

  56. Chan, T. K., Todd, D., Lai, M. C. S. Glucose 6-phosphate dehydrogenase: identity of erythrocyte and leukocyte enzyme with report of a new variant in Chinese. Biochem. Genet. 6: 119-124, 1972. [PubMed: 4666752, related citations] [Full Text]

  57. Chan, T. K., Todd, D. Characteristics and distribution of glucose-6-phosphate dehydrogenase-deficient variants in South China. Am. J. Hum. Genet. 24: 475-484, 1972. [PubMed: 5031985, related citations]

  58. Chang, J.-G., Chiou, S.-S., Perng, L.-I., Chen, T.-C., Liu, T.-C., Lee, L.-S., Chen, P.-H., Tang, T. K. Molecular characterization of glucose-6-phosphate dehydrogenase (G6PD) deficiency by natural and amplification created restriction sites: five mutations account for most G6PD deficiency cases in Taiwan. Blood 80: 1079-1082, 1992. [PubMed: 1323345, related citations]

  59. Chen, E. Y., Cheng, A., Lee, A., Kuang, W.-J., Hillier, L., Green, P., Schlessinger, D., Ciccodicola, A., D'Urso, M. Sequence of human glucose-6-phosphate dehydrogenase cloned in plasmids and a yeast artificial chromosome. Genomics 10: 792-800, 1991. [PubMed: 1889820, related citations] [Full Text]

  60. Chen, H.-L., Huang, M.-J., Huang, C.-S., Tang, T. K. G6PD NanKang (517 T-to-C; 173 phe-to-leu): a new Chinese G6PD variant associated with neonatal jaundice. Hum. Hered. 46: 201-204, 1996. [PubMed: 8807322, related citations] [Full Text]

  61. Cherniak, N. B., Batischev, A. I., Lanzina, N. V., Tokarev, Y. N., Alexeyev, G. A. Electrophoretic and kinetic properties of glucose-6-phosphate dehydrogenase from erythrocytes or patients with hemolytic anemia, related to deficiency of the enzyme activity. Vopr. Med. Khim. 23: 166-171, 1977.

  62. Childs, B., Zinkham, W., Browne, E. A., Kimbro, E. L., Torbert, J. V. A genetic study of a defect in glutathione metabolism of the erythrocytes. Bull. Johns Hopkins Hosp. 102: 21-37, 1958. [PubMed: 13500095, related citations]

  63. Chiu, D. T. Y., Zuo, L., Chao, L., Chen, E., Louie, E., Lubin, B., Liu, T. Z., Du, C. S. Molecular characterization of glucose-6-phosphate dehydrogenase (G6PD) deficiency in patients of Chinese descent and identification of new base substitutions in the human G6PD gene. Blood 81: 2150-2154, 1993. [PubMed: 8471773, related citations]

  64. Chockkalingam, K., Board, P. G., Breguet, G. Glucose-6-phosphate dehydrogenase variants of Bali Island (Indonesia). Hum. Genet. 60: 60-62, 1982. [PubMed: 7076249, related citations] [Full Text]

  65. Chockkalingam, K., Board, P. G., Nurse, G. T. Glucose-6-phosphate dehydrogenase deficiency in Papua New Guinea: the description of 13 new variants. Hum. Genet. 60: 189-192, 1982. [PubMed: 7076260, related citations] [Full Text]

  66. Chockkalingam, K., Board, P. G. Further evidence for heterogeneity of glucose-6-phosphate dehydrogenase deficiency in Papua New Guinea. Hum. Genet. 56: 209-212, 1980. [PubMed: 7450778, related citations] [Full Text]

  67. Cianfriglia, M., Miggiano, V. C., Meo, T., Muller, H. J., Muller, E., Battistuzzi, G. Evidence for synteny between the rabbit gene loci coding for HPRT, PGK and G6PD in mouse-rabbit somatic cell hybrids. (Abstract) Cytogenet. Cell Genet. 25: 142, 1979.

  68. Cohn, J., Carter, N., Warburg, M. Glucose-6-phosphate dehydrogenase deficiency in a native Danish family: a new variant. Scand. J. Haemat. 23: 403-406, 1979. [PubMed: 542803, related citations] [Full Text]

  69. Colonna-Romano, S., Iolascon, A., Lippo, S., Pinto, L., Cutillo, S., Battistuzzi, G. Genetic heterogeneity at the glucose-6-phosphate dehydrogenase locus in southern Italy: a study on the population of Naples. Hum. Genet. 69: 228-232, 1985. [PubMed: 3980015, related citations] [Full Text]

  70. Cooper, D. W., Johnston, P. G., Murtagh, C. E., Sharman, G. B., Vandeberg, J. L., Poole, W. E. Sex-linked isozymes and sex-chromosome evolution and inactivation in kangaroos. In: Markert, C. L. (ed.): Isozymes. Vol. 3. Developmental Biology. New York: Academic Press (pub.) 1975. Pp. 559-573.

  71. Corash, L., Spielberg, S., Bartsocas, C., Boxer, L., Steinherz, R., Sheetz, M., Egan, M., Schlessleman, J., Schulman, J. D. Reduced chronic hemolysis during high-dose vitamin E administration in Mediterranean-type glucose-6-phosphate dehydrogenase deficiency. New Eng. J. Med. 303: 416-420, 1980. [PubMed: 7393270, related citations] [Full Text]

  72. Corcoran, C. M., Calabro, V., Tamagnini, G., Town, M., Haidar, B., Vulliamy, T. J., Mason, P. J., Luzzatto, L. Molecular heterogeneity underlying the G6PD Mediterranean phenotype. Hum. Genet. 88: 688-690, 1992. [PubMed: 1551674, related citations] [Full Text]

  73. Costa, E., Cabeda, J. M., Vieira, E., Pinto, R., Pereira, S. A., Ferraz, L., Santos, R., Barbot, J. Glucose-6-phosphate dehydrogenase Aveiro: a de novo mutation associated with chronic nonspherocytic hemolytic anemia. Blood 95: 1499-1501, 2000. [PubMed: 10666231, related citations]

  74. Crookston, J. H., Yoshida, A., Lin, M., Booser, D. J. G6PD Toronto. Biochem. Genet. 8: 259-265, 1973. [PubMed: 4701992, related citations] [Full Text]

  75. Csepreghy, M., Hall, M. K., Berkow, R. L., Jackson, S., Prchal, J. T. Characterization of a new G6PD variant: G6PD Titusville. Am. J. Med. Sci. 297: 114-117, 1989. [PubMed: 2919631, related citations] [Full Text]

  76. Csepreghy, M., Yeilding, A., Lilly, M., Hall, K., Scott, C. W., Prchal, J. T. Characterization of a new glucose-6-phosphate dehydrogenase variant: G6PD Central City. Am. J. Hemat. 28: 61-62, 1988. [PubMed: 3369438, related citations] [Full Text]

  77. D'Urso, M., Luzzatto, L., Perroni, L., Ciccodicola, A., Gentile, G., Peluso, I., Persico, M. G., Pizzella, T., Toniolo, D., Vulliamy, T. J. An extensive search for RFLP in the human glucose-6-phosphate dehydrogenase locus has revealed a silent mutation in the coding sequence. Am. J. Hum. Genet. 42: 735-741, 1988. [PubMed: 2895981, related citations]

  78. Davidson, R. G., Nitowsky, H. M., Childs, B. Demonstration of two populations of cells in the human female heterozygous for glucose-6-phosphate dehydrogenase variants. Proc. Nat. Acad. Sci. 50: 481-485, 1963. [PubMed: 14067093, related citations] [Full Text]

  79. De Flora, A., Morelli, A., Benatti, U., Giuntini, P., Ferraris, A. M., Galiano, S., Ravazzolo, R., Gaetani, G. F. G6PD Napoli and Ferrara II: two new glucose-6-phosphate dehydrogenase variants having similar characteristics but different intracellular lability and specific activity. Brit. J. Haemat. 48: 417-423, 1981. [PubMed: 7259990, related citations] [Full Text]

  80. De Vita, G., Alcalay, M., Sampietro, M., Cappelini, M. D., Fiorelli, G., Toniolo, D. Two point mutations are responsible for G6PD polymorphism in Sardinia. Am. J. Hum. Genet. 44: 233-240, 1989. [PubMed: 2912069, related citations]

  81. Dern, R. J., McCurdy, P. R., Yoshida, A. A new structural variant of glucose-6-phosphate dehydrogenase with a high production rate (G6PD Hektoen). J. Lab. Clin. Med. 73: 283-290, 1969. [PubMed: 4974311, related citations]

  82. Du, C. S., Xu, Y. K., Hua, X. Y., Wu, Q. L., Liu, L. B. Glucose-6-phosphate dehydrogenase variants and their frequency in Guangdong, China. Hum. Genet. 80: 385-388, 1988. [PubMed: 3198117, related citations] [Full Text]

  83. Du, C. S. Studies on erythrocyte glucose-6-phosphate dehydrogenase variants in Chinese. I. Gd(-) Lizu-Baisha. Chin. Med. J. 61: 729-732, 1981. [PubMed: 6805883, related citations]

  84. Du, C., Xu, Y., Hua, X., Wu, Q., Liu, L., Wu, M. Studies on erythrocyte glucose-6-phosphate dehydrogenase variants in Chinese. III: Gd(-) Miaozu-Baisha. Acta Genet. Sin. 11: 153-158, 1984. [PubMed: 6805883, related citations]

  85. Du, C.-S., Hua, X.-Y., Wu, Q.-L., Li, C.-Q., Zheng, J.-F., Li, H.-L. Studies on erythrocyte glucose-6-phosphate dehydrogenase variants in Chinese. IV. Gd(-) Gaohe associated with paroxysmal nocturnal hemoglobinuria. Chinese J. Pathophysiol. 1: 12-15, 1985.

  86. Eber, S. W., Gahr, M., Schroter, W. Glucose-6-phosphate dehydrogenase (G6PD) Iserlohn and G6PD Regensburg: two new severe enzyme defects in German families. Blut 51: 109-115, 1985. [PubMed: 3849316, related citations] [Full Text]

  87. Echard, G., Gillois, M. G6PD--alpha-GAL-PGK--HPRT synteny in the rabbit, Oryctolagus cunniculus. (Abstract) Cytogenet. Cell Genet. 25: 148-149, 1979.

  88. Efferth, T., Bachli, E. B., Schwarzl, S. M., Goede, J. S., West, C., Smith, J. C., Beutler, E. Glucose-6-phosphate dehydrogenase (G6PD) deficiency-type Zurich: a splice site mutation as an uncommon mechanism producing enzyme deficiency. (Letter) Blood 104: 2608 only, 2004. [PubMed: 15466166, related citations] [Full Text]

  89. Elizondo, J., Saenz, G. F., Paez, C. A., Ramon, M., Garcia, M., Gutierrez, A., Estrada, M. G6PD Puerto Limon: a new deficient variant of glucose-6-phosphate dehydrogenase associated with congenital nonspherocytic hemolytic anemia. Hum. Genet. 62: 110-112, 1982. [PubMed: 7160841, related citations] [Full Text]

  90. Engstrom, P. F., Beutler, E. G-6-PD Tripler: a unique variant associated with chronic hemolytic disease. Blood 36: 10-13, 1970. [PubMed: 5421739, related citations]

  91. Epstein, C. J. Mammalian oocytes: X-chromosome activity. Science 163: 1078-1079, 1969. [PubMed: 5764873, related citations] [Full Text]

  92. Ermakov, N. V., Chernyak, N. B., Tokarev, Y. N. Properties of new variant of glucose-6-phosphate dehydrogenase (Regar variant): glucose metabolism in erythrocytes containing abnormal enzyme. Biokhimiia 48: 577-583, 1983.

  93. Estrada, M., Garcia, M., Gutierrez, A., Quintero, I., Gonzalez, R. G6PD Varadero. Vox Sang. 43: 102-104, 1982. [PubMed: 7123903, related citations] [Full Text]

  94. Fairbanks, V. F., Nepo, A. G., Beutler, E., Dickson, E. R., Honig, G. Glucose-6-phosphate dehydrogenase variants: reexamination of G6PD Chicago and Cornell and a new variant (G6PD Pea Ridge) resembling G6PD Chicago. Blood 55: 216-220, 1980. [PubMed: 7353072, related citations]

  95. Faust, C. J., Levinson, B., Gitschier, J., Herman, G. E. Extension of the physical map in the region of the mouse X chromosome homologous to human Xq28 and identification of an exception to conserved linkage. Genomics 13: 1289-1295, 1992. [PubMed: 1354645, related citations] [Full Text]

  96. Feldman, R., Gromisch, D. S., Luhby, A. L., Beutler, E. Congenital nonspherocytic hemolytic anemia due to glucose-6-phosphate dehydrogenase East Harlem: a new deficient variant. J. Pediat. 90: 89-91, 1977. [PubMed: 830903, related citations] [Full Text]

  97. Fernandez, M., Fairbanks, V. F. Glucose-6-phosphate dehydrogenase deficiency in the Philippines: report of a new variant--G6PD Panay. Mayo Clin. Proc. 43: 645-660, 1968. [PubMed: 5710590, related citations]

  98. Filosa, E. Personal Communication. La Jolla, Calif. 1989.

  99. Filosa, S., Calabro, V., Vallone, D., Poggi, V., Mason, P., Pagnini, D., Alfinito, F., Rotoli, B., Martini, G., Luzzatto, L., Battistuzzi, G. Molecular basis of chronic non-spherocytic haemolytic anaemia: a new G6PD variant (393arg-to-his) with abnormal K(m) G6P and marked in vivo instability. Brit. J. Haemat. 80: 111-116, 1992. [PubMed: 1536798, related citations] [Full Text]

  100. Filosa, S., Giacometti, N., Wangwei, C., De Mattia, D., Pagnini, D., Alfinito, F., Schettini, F., Luzzatto, L., Martini, G. Somatic-cell selection is a major determinant of the blood-cell phenotype in heterozygotes for glucose-6-phosphate dehydrogenase mutations causing severe enzyme deficiency. Am. J. Hum. Genet. 59: 887-895, 1996. [PubMed: 8808605, related citations]

  101. Fiorelli, G., Manoussakis, C., Sampietro, M., Pittalis, S., Guglielmino, C. R., Cappellini, M. D. Different polymorphic variants of glucose-6-phosphate dehydrogenase (G6PD) in Italy. Ann. Hum. Genet. 53: 229-236, 1989. [PubMed: 2596828, related citations] [Full Text]

  102. Fite, E., Morell, F., Zuazu, J., Julia, A., Morera, J. Leucocyte glucose-6-phosphate dehydrogenase deficiency and necrotizing pneumonia. Europ. J. Resp. Dis. 64: 150-154, 1983. [PubMed: 6832252, related citations]

  103. Francke, U., Bakay, B., Connor, J. D., Coldwell, J. G., Nyhan, W. L. Linkage relationships of X-linked enzymes glucose-6-phosphate dehydrogenase and hypoxanthine guanine phosphoribosyltransferase. Am. J. Hum. Genet. 26: 512-522, 1974. [PubMed: 4842775, related citations]

  104. Friedman, M. J., Trager, W. The biochemistry of resistance to malaria. Sci. Am. 244(3): 154-164, 1981. [PubMed: 6163210, related citations] [Full Text]

  105. Frigerio, R., Sole, G., Olla, N., Lovicu, M., Passiu, G., Carcassi, U. Cagliari II: a new G-6-PD variant. Haematologica 72: 241-243, 1987. [PubMed: 3114091, related citations]

  106. Fujii, H., Miwa, S., Takegawa, S., Takahashi, K., Hirono, A., Takizawa, T., Morisaki, T., Kanno, H., Taguchi, T., Okamura, J. Gd(-) Gifu and Gd(-) Fukuoka: two new variants of glucose-6-phosphate dehydrogenase found in Japan. Hum. Genet. 66: 276-278, 1984. [PubMed: 6714986, related citations] [Full Text]

  107. Fujii, H., Miwa, S., Tani, K., Takegawa, S., Fujinami, N., Takahashi, K., Nakayama, S., Konno, M., Sato, T. Glucose 6-phosphate dehydrogenase variants: a unique variant (G6PD Kobe) showed an extremely increased affinity for galactose 6-phosphate and a new variant (G6PD Sapporo) resembling G6PD Pea Ridge. Hum. Genet. 58: 405-407, 1981. [PubMed: 7327562, related citations] [Full Text]

  108. Fusco, F., Paciolla, M., Napolitano, F., Pescatore, A., D'Addario, I., Bal, E., Lioi, M. B., Smahl, A., Miano, M. G., Ursini, M. V. Genomic architecture at the incontinentia pigmenti locus favours de novo pathological alleles through different mechanisms. Hum. Molec. Genet. 21: 1260-1271, 2012. [PubMed: 22121116, related citations] [Full Text]

  109. Gaetani, G. F., Galiano, S., Melani, C., Miglino, M., Forni, G. L., Napoli, G., Perrone, L., Ferraris, A. M. A new glucose-6-phosphate dehydrogenase variant with congenital nonspherocytic hemolytic anemia (G6PD Genova): biochemical characterization and mosaicism expression in the heterozygote. Hum. Genet. 84: 337-340, 1990. [PubMed: 2307454, related citations] [Full Text]

  110. Gahr, M., Bornhalm, D., Schroter, W. Haemolytic anemia due to glucose 6-phosphate dehydrogenase (G6PD) deficiency: demonstration of two new biochemical variants, G6PD Hamm and G6PD Tarsus. Brit. J. Haemat. 33: 363-370, 1976. [PubMed: 1276080, related citations] [Full Text]

  111. Gahr, M., Bornhalm, D., Schroter, W. Biochemische Eigenschaften einer neuen Variante des Glucose-6-phosphadehydrogenase (G6PD) Mangels mit Favismus: G6PD Bielefeld. Klin. Wschr. 55: 379-384, 1977. [PubMed: 859288, related citations] [Full Text]

  112. Gahr, M., Schroter, W., Sturzenegger, M., Bornhalm, D., Marti, H. R. Glucose 6-phosphate dehydrogenase (G6PD) deficiency in Switzerland. Helv. Paediat. Acta 31: 159-166, 1976. [PubMed: 955939, related citations]

  113. Gahr, M., Schroter, W. Glucose 6-phosphate dehydrogenase (G6PD) Hamburg, a new variant with chronic nonspherocytic hemolytic anemia. Europ. J. Clin. Invest. 4: 187-191, 1974. [PubMed: 4838696, related citations] [Full Text]

  114. Geerdink, R. A., Horst, R., Staal, G. E. An Iraqi Jewish family with a new red cell glucose 6-phosphate dehydrogenase variant (Gd-Bagdad) and kernicterus. Isr. J. Med. Sci. 9: 1040-1043, 1973. [PubMed: 4753845, related citations]

  115. Gellin, J., Benne, F., Renard, C., Vaiman, M., Hors-Cayla, M. C., Gillois, M. Pig gene mapping: synteny, attempt to assign the histocompatibility complex (SLA). (Abstract) Cytogenet. Cell Genet. 25: 159, 1979.

  116. Gomez-Gallego, F., Garrido-Pertierra, A., Bautista, J. M. Structural defects underlying protein dysfunction in human glucose-6-phosphate dehydrogenase A- deficiency. J. Biol. Chem. 275: 9256-9262, 2000. [PubMed: 10734064, related citations] [Full Text]

  117. Gonzalez, R., Estrada, M., Garcia, M., Gutierrez, A. G6PD Ciudad de la Habana: a new slow variant with deficiency found in a Cuban family. Hum. Genet. 55: 133-135, 1980. [PubMed: 7450750, related citations] [Full Text]

  118. Gonzalez, R., Wade, M., Estrada, M., Svarch, E., Colombo, B. G6PD Pinar del Rio: a new variant discovered in a Cuban family. Biochem. Genet. 15: 909-913, 1977. [PubMed: 588239, related citations] [Full Text]

  119. Goss, S. J., Harris, H. Gene transfer by means of cell fusion. I. Statistical mapping of the human X-chromosome by analysis of radiation induced gene segregation. J. Cell Sci. 25: 17-37, 1977. [PubMed: 561093, related citations] [Full Text]

  120. Gourdin, D., Vergnes, H., Bouloux, C., Ruffie, J., Gherardi, M. Polymorphism of erythrocyte G6PD in the baboon. Am. J. Phys. Anthrop. 37: 281-288, 1972. [PubMed: 4628608, related citations] [Full Text]

  121. Gray, G. R., Stamatoyannopoulos, G., Naiman, S. C., Kliman, M. R., Klebanoff, S. J., Austin, T., Yoshida, A., Robinson, G. C. G. Neutrophil dysfunction, chronic granulomatous disease, and non-spherocytic haemolytic anaemia caused by complete deficiency of glucose-6-phosphate dehydrogenase. Lancet 302: 530-534, 1973. Note: Originally Volume II. [PubMed: 4125296, related citations] [Full Text]

  122. Grossman, A., Ramanathan, K., Justice, P., Gordon, J., Shahidi, N. T., Hsia, D. Y. Y. Congenital nonspherocytic hemolytic anemia associated with erythrocyte G-6-PD deficiency in a Negro family. Pediatrics 37: 624-629, 1966. [PubMed: 5930024, related citations]

  123. Gutierrez, A., Garcia, M., Estrada, M., Quintero, I., Gonzalez, R. Glucose-6-phosphate dehydrogenase (G6PD) Guantanamo and G6PD Caujeri: two new glucose-6-phosphate dehydrogenase-deficient variants found in Cuba. Biochem. Genet. 25: 231-238, 1987. [PubMed: 3606560, related citations] [Full Text]

  124. Hall, K., Schreeder, M. T., Prchal, J. T. G6PD Huntsville: a new glucose-6-phosphate dehydrogenase associated with chronic hemolytic anemia. Hum. Genet. 79: 90-91, 1988. [PubMed: 3366466, related citations] [Full Text]

  125. Harkonen, M., Vuopio, P. Red cell glucose-6-phosphate dehydrogenase deficiency in Finland. Ann. Clin. Res. 6: 187-197, 1974. [PubMed: 4154719, related citations]

  126. Harley, J. D., Agar, N. S., Yoshida, A. Glucose 6-phosphate dehydrogenase variant Gd(+) Alexandra associated with neonatal jaundice and Gd(-) Camperdown in a young man with lamellar cataracts. J. Lab. Clin. Med. 91: 295-300, 1978. [PubMed: 23402, related citations]

  127. Helge, H., Borner, K. Kongenitale nichtsphaerozytare haemolytische Anaemie, Katarakt und Glucose-6-phosphat-dehydrogenase-mangel. Dtsch. Med. Wschr. 91: 1584-1589, 1966. [PubMed: 4380311, related citations] [Full Text]

  128. Hirono, A., Beutler, E. Molecular cloning and nucleotide sequence of cDNA for human glucose-6-phosphate dehydrogenase variant A(-). Proc. Nat. Acad. Sci. 85: 3951-3954, 1988. [PubMed: 2836867, related citations] [Full Text]

  129. Hirono, A., Beutler, E. Alternative splicing of human glucose-6-phosphate dehydrogenase messenger RNA in different tissues. J. Clin. Invest. 83: 343-346, 1989. [PubMed: 2910917, related citations] [Full Text]

  130. Hirono, A., Fujii, H., Shima, M., Miwa, S. G6PD Nara: a new class 1 glucose-6-phosphate dehydrogenase variant with an eight amino acid deletion. Blood 82: 3250-3252, 1993. [PubMed: 8241497, related citations]

  131. Hirono, A., Kawate, K., Honda, A., Fujii, H., Miwa, S. A single mutation 202G-A in the human glucose-6-phosphate dehydrogenase gene (G6PD) can cause acute hemolysis by itself. (Letter) Blood 99: 1498 only, 2002. [PubMed: 11852882, related citations] [Full Text]

  132. Hirono, A., Kuhl, W., Gelbart, T., Forman, L., Fairbanks, V. F., Beutler, E. Identification of the binding domain for NADP(+) of human glucose-6-phosphate dehydrogenase by sequence analysis of mutants. Proc. Nat. Acad. Sci. 86: 10015-10017, 1989. [PubMed: 2602358, related citations] [Full Text]

  133. Hitzeroth, H. W., Bender, K. Age-dependency of somatic selection in South African Negro G-6-PD heterozygotes. Hum. Genet. 58: 338-343, 1981. [PubMed: 7327555, related citations] [Full Text]

  134. Honig, G. R., Habacon, E., Vida, L. N., Matsumoto, F., Beutler, E. Three new variants of glucose-6-phosphate dehydrogenase associated with chronic nonspherocytic hemolytic anemia: G6PD Lincoln Park, G6PD Arlington Heights, and G6PD West Town. Am. J. Hemat. 6: 353-360, 1979. [PubMed: 532804, related citations] [Full Text]

  135. Hook, E. B., Stamatoyannopoulos, G., Yoshida, A., Motulsky, A. G. Glucose-6-phosphate dehydrogenase Madrona: a slow electrophoretic glucose-6-phosphate dehydrogenase variant with kinetic characteristics similar to those of normal type. J. Lab. Clin. Med. 72: 404-409, 1968. [PubMed: 5674071, related citations]

  136. Hors-Cayla, M. C., Heuertz, S., Van Cong, N., Benne, F. Cattle gene mapping by somatic cell hybridization. (Abstract) Cytogenet. Cell Genet. 25: 165-166, 1979.

  137. Howell, E. B., Nelson, A. J., Jones, O. W. A new G-6-PD variant associated with chronic non-spherocytic haemolytic anaemia in a Negro family. J. Med. Genet. 9: 160-164, 1972. [PubMed: 5046624, related citations] [Full Text]

  138. Hutz, M. H., Yoshida, A., Salzano, F. M. Three rare G-6-PD variants from Porto Alegre, Brazil. Hum. Genet. 39: 191-197, 1977. [PubMed: 598829, related citations] [Full Text]

  139. Iancovici-Kidon, M., Sthoeger, D., Abrahamov, A., Wolach, B., Beutler, E., Gelbart, T., Barak, Y. A new exon 9 glucose-6-phosphate dehydrogenase mutation (G6PD 'Rehovot') in a Jewish Ethiopian family with variable phenotypes. Blood Cells Molec. Dis. 26: 567-571, 2000. Note: Erratum: Blood Cells Molec. Dis. 27: 351 only, 2001. [PubMed: 11112389, related citations] [Full Text]

  140. Ishwad, C. S., Naik, S. N. A new glucose-6-phosphate dehydrogenase variant (G-6-PD Kalyan) found in a Koli family. Hum. Genet. 66: 171-175, 1984. [PubMed: 6714978, related citations] [Full Text]

  141. Iwai, K., Hirono, A., Matsuoka, H., Kawamoto, F., Horie, T., Lin, K., Tantular, I. S., Dachlan, Y. P., Notopuro, H., Hidayah, N. I., Salim, A. M. A., Fujii, H., Miwa, S., Ishii, A. Distribution of glucose-6-phosphate dehydrogenase mutations in Southeast Asia. Hum. Genet. 108: 445-449, 2001. [PubMed: 11499668, related citations] [Full Text]

  142. Jain, M., Cui, L., Brenner, D. A., Wang, B., Handy, D. E., Leopold, J. A., Loscalzo, J., Apstein, C. S., Liao, R. Increased myocardial dysfunction after ischemia-reperfusion in mice lacking glucose-6-phosphate dehydrogenase. Circulation 109: 898-903, 2004. [PubMed: 14757696, related citations] [Full Text]

  143. Jiang, W., Yu, G., Liu, P., Geng, Q., Chen, L., Lin, Q., Ren, X., Ye, W., He, Y., Guo, Y., Duan, S., Wen, J., and 9 others. Structure and function of glucose-6-phosphate dehydrogenase-deficient variants in Chinese population. Hum. Genet. 119: 463-478, 2006. [PubMed: 16607506, related citations] [Full Text]

  144. Johnson, G. J., Kaplan, M. E., Beutler, E. G6PD Long Prairie: a new mutant exhibiting normal sensitivity to inhibition by NADPH and accompanied by nonspherocytic hemolytic anemia. Blood 49: 247-251, 1977. [PubMed: 12846, related citations]

  145. Johnston, P. G., VandeBerg, J. L., Sharman, G. B. Inheritance of erythrocyte glucose 6-phosphate dehydrogenase in the red-necked wallaby, Macropus rufogriseus (Desmarest) consistent with paternal X inactivation. Biochem. Genet. 13: 235-242, 1975. [PubMed: 1147889, related citations] [Full Text]

  146. Junien, C., Kaplan, J. C., Meienhofer, M. C., Maigret, P., Sender, A. G6PD Baudelocque: a new unstable variant characterized in cultured fibroblasts. Enzyme 18: 45-59, 1974. [PubMed: 4852206, related citations]

  147. Kaeda, J. S., Chhotray, G. P., Ranjit, M. R., Bautista, J. M., Reddy, P. H., Stevens, D., Naidu, J. M., Britt, R. P., Vulliamy, T. J., Luzzatto, L., Mason, P. J. A new glucose-6-phosphate dehydrogenase variant, G6PD Orissa (44 ala-gly), is the major polymorphic variant in tribal populations in India. Am. J. Hum. Genet. 57: 1335-1341, 1995. [PubMed: 8533762, related citations]

  148. Kageoka, T., Satoh, C., Goriki, K., Fujita, M., Neriishi, S., Yamamura, K., Kaneko, J., Masunari, N. Electrophoretic variants of blood proteins in Japanese. IV. Prevalence and enzymologic characteristics of G6PD variants in Hiroshima and Nagasaki. Hum. Genet. 70: 101-108, 1985. [PubMed: 4007855, related citations] [Full Text]

  149. Kahn, A., Bernard, J.-F., Cottreau, D., Mazie, J., Boivin, P. Gd(-) Abrami, a deficient G6PD variant with hemizygous expression in blood cells of a woman with primary myelofibrosis. Humangenetik 30: 41-46, 1975. [PubMed: 241700, related citations] [Full Text]

  150. Kahn, A., Boivin, P., Hakim, J., Lagneau, J. Heterogeneite des glucose-6-phosphate dehydrogenase erythrocytaire deficitaires dans la race noire: etude cinetique et description de deux nouvelles variants Gd(-) Dakar et Gd(-) Mali. Nouv. Rev. Franc. Hemat. 11: 741-758, 1971. [PubMed: 4261190, related citations]

  151. Kahn, A., Boivin, P., Lagneau, J. Phenotypes de la glucose-6-phosphate dehydrogenase erythrocytaire dans la race noire. Humangenetik 18: 261-270, 1973. [PubMed: 4719636, related citations] [Full Text]

  152. Kahn, A., Boulard, M., Hakim, J., Schaison, G., Boivin, P., Bernard, J. Anemie hemolytique congenitale non spherocytaire par deficit en glucose 6-phosphate dehydrogenase erythrocytaire: description de deux nouvelles variants: Gd(-) Saint Louis (Paris) et Gd(-) Hayem. Nouv. Rev. Franc. Hemat. 14: 587-600, 1974. [PubMed: 4141762, related citations]

  153. Kahn, A., Dao, C., Cottreau, D., Bilski-Pasquier, G. 'Gd(-) Hotel Dieu': a new G-6PD variant with chronic hemolysis in a Negro patient from Senegal. Hum. Genet. 39: 353-358, 1977. [PubMed: 598844, related citations] [Full Text]

  154. Kahn, A., Esters, A., Habedank, M. Gd(-) Aachen, a new variant of deficient glucose-6-phosphate dehydrogenase. Hum. Genet. 32: 171-180, 1976. [PubMed: 1270075, related citations] [Full Text]

  155. Kahn, A., Hakim, J., Cottreau, D., Boivin, P. Gd (-) Matam. An African glucose 6-phosphate dehydrogenase variant with enzyme deficiency. Biochemical and immunological properties in various hemopoietic tissues. Clin. Chim. Acta 59: 183-190, 1975. [PubMed: 235389, related citations] [Full Text]

  156. Kahn, A., North, M. L., Cottreau, D., Giron, G., Lang, J. M. G6PD Vientiane: a new glucose-6-phosphate dehydrogenase variant with increased stability. Hum. Genet. 43: 85-89, 1978. [PubMed: 27443, related citations] [Full Text]

  157. Kahn, A., North, M. L., Messer, J., Boivin, P. G-6PD 'Ankara': a new G-6PD variant with deficiency found in a Turkish family. Humangenetik 27: 247-250, 1975. [PubMed: 1150245, related citations] [Full Text]

  158. Kanno, H., Huang, I.-Y., Kan, Y. W., Yoshida, A. Two structural genes on different chromosomes are required for encoding the major subunit of human red cell glucose-6-phosphate dehydrogenase. Cell 58: 595-606, 1989. [PubMed: 2758468, related citations] [Full Text]

  159. Kanno, H., Takano, T., Fujii, H., Tani, K., Morisaki, T., Hirono, A., Kumakawa, T., Ogura, H., Takahashi, K., Tsutsumi, H., Miwa, S. A new glucose-6-phosphate variant (G6PD Iwate) associated with congenital non-spherocytic hemolytic anemia. Acta Haemat. Jpn. 51: 715-719, 1987.

  160. Kaplan, J. C., Hanzlickova-Leroux, A., Nicholas, A. M., Rosa, R., Weiler, C., Lepercq, G. A new glucose-6-phosphate dehydrogenase variant (G6PD Port-Royal). Enzyme 12: 25-32, 1971. [PubMed: 5105366, related citations] [Full Text]

  161. Kaplan, J. C., Rosa, R., Seringe, P., Hoeffel, J. C. Le polymorphisme genetique de la glucose-6-phosphate deshydrogenase erythrocytaire chez l'homme. II. Etude d'une nouvelle variete a activite diminuee: le type 'Kabyle'. Enzymol. Biol. Clin. (Basel) 8: 332-340, 1967. [PubMed: 5299577, related citations]

  162. Kaplan, M., Hammerman, C., Vreman, H. J., Stevenson, D. K., Beutler, E. Acute hemolysis and severe neonatal hyperbilirubinemia in glucose-6-phosphate dehydrogenase deficient heterozygotes. J. Pediat. 139: 137-140, 2001. [PubMed: 11445808, related citations] [Full Text]

  163. Kaplan, M., Renbaum, P., Levy-Lahad, E., Hammerman, C., Lahad, A., Beutler, E. Gilbert syndrome and glucose-6-phosphate dehydrogenase deficiency: a dose-dependent genetic interaction crucial to neonatal hyperbilirubinemia. Proc. Nat. Acad. Sci. 94: 12128-12132, 1997. [PubMed: 9342374, images, related citations] [Full Text]

  164. Karadsheh, N. S., Awidi, A. S., Tarawneh, M. S. Two new glucose-6 phosphate dehydrogenase (G6PD) variants associated with hemolytic anemia: G6PD Amman-1 and G6PD Amman-2. Am. J. Hemat. 22: 185-192, 1986. [PubMed: 3706292, related citations] [Full Text]

  165. Kay, A. C., Kuhl, W., Prchal, J., Beutler, E. The origin of glucose-6-phosphate-dehydrogenase (G6PD) polymorphisms in African-Americans. Am. J. Hum. Genet. 50: 394-398, 1992. [PubMed: 1734718, related citations]

  166. Kirkman, H. N., Eng, L.-I. L. Variants of glucose 6-phosphate dehydrogenase in Indonesia. Nature 221: 959, 1969. [PubMed: 5765510, related citations] [Full Text]

  167. Kirkman, H. N., Kidson, C., Kennedy, M. Variants of human glucose-6-phosphate dehydrogenase. Studies of samples from New Guinea. In: Beutler, E.: Hereditary Disorders of Erythrocyte Metabolism. New York: Grune and Stratton (pub.) 1968. Pp. 126-145.

  168. Kirkman, H. N., McCurdy, P. R., Naiman, J. L. Functionally abnormal glucose-6-phosphate dehydrogenases. Cold Spring Harbor Symp. Quant. Biol. 29: 391-398, 1964. [PubMed: 14278484, related citations] [Full Text]

  169. Kirkman, H. N., Ramot, B., Lee, J. T. Altered aggregational properties in a genetic variant of human glucose-6-phosphate dehydrogenase. Biochem. Genet. 3: 137-150, 1969. [PubMed: 5364922, related citations] [Full Text]

  170. Kirkman, H. N., Riley, H. D., Jr. Congenital nonspherocytic hemolytic anemia. Am. J. Dis. Child. 102: 313-320, 1961. [PubMed: 13756265, related citations] [Full Text]

  171. Kirkman, H. N., Rosenthal, I. M., Simon, E. B., Carson, P. E., Brinson, A. G. 'Chicago I' variant of glucose-6-phosphate dehydrogenase in congenital hemolytic disease. J. Lab. Clin. Med. 63: 715-725, 1964. [PubMed: 14158057, related citations]

  172. Kirkman, H. N., Schettini, E., Pickard, B. M. Mediterranean variant of glucose-6-phosphate dehydrogenase. J. Lab. Clin. Med. 63: 726-735, 1964.

  173. Kirkman, H. N., Simon, E. R., Pickard, B. M. Seattle variant of glucose-6-phosphate dehydrogenase. J. Lab. Clin. Med. 66: 834-840, 1965. [PubMed: 5844610, related citations]

  174. Kissin, C., Cotte, J. Etude d'un variant de glucose-6-phosphate deshydrogenase: I B type Constantine. Enzymol. Biol. Clin. (Basel) 11: 277-284, 1970. [PubMed: 5414134, related citations] [Full Text]

  175. Kitao, T., Ito, K., Hattori, K., Matsuki, T., Yoneyama, Y. G6PD Kanazawa: a new variant of glucose-6-phosphate dehydrogenase associated with congenital nonspherocytic hemolytic anemia. Acta Haemat. 68: 131-135, 1982. [PubMed: 6812368, related citations] [Full Text]

  176. Kojima, H. Congenital nonspherocytic hemolytic disease (CNHD) due to a G-6-PD variant: G-6-PD Kyoto. Acta Haemat. Jpn. 35: 32-38, 1972. [PubMed: 4673853, related citations]

  177. Koliakos, G., Kalomenopoulou, M., Grammatikos, P., Dimitriadou, A., Kouzi-Koliakos, K., Zacharaki, R., Skaragas, G., Kokka, A., Trakatellis, A. A new glucose 6-phosphate dehydrogenase variant (G6PD Thessaloniki) in a patient with idiopathic myelofibrosis. Hum. Hered. 39: 141-149, 1989. [PubMed: 2591977, related citations] [Full Text]

  178. Krasnopolskaya, K. D., Bochkov, N. P. Genetic heterogeneity of hereditary enzymopathies. Vestn. Akad. Med. Nauk. SSSR 9: 56-64, 1982. [PubMed: 7148122, related citations]

  179. Krasnopolskaya, K. D., Shatskaya, T. L., Filippov, I. K., Annenkov, G. A., Zakharova, T. V., Mekhtiev, N. K., Movsum-Zade, K. M. Genetic heterogeneity of G6PD deficiency: study of mutant alleles in Shekii district of Azerbaijan. Genetika 13: 1455-1461, 1977. [PubMed: 144081, related citations]

  180. Kumakawa, T., Suzuki, S., Fujii, H., Miwa, S. Frequency of glucose 6-phosphate dehydrogenase (G6PD) deficiency in Tokyo and a new variant: G6PD Musashino. Acta Haemat. Jpn. 50: 25-28, 1987. [PubMed: 3591235, related citations]

  181. Kurdi-Haidar, B., Mason, P. J., Berrebi, A., Ankra-Badu, G., Al-Ali, A., Oppenheim, A., Luzzatto, L. Origin and spread of the glucose-6-phosphate dehydrogenase variant (G6PD-Mediterranean) in the Middle East. Am. J. Hum. Genet. 47: 1013-1019, 1990. [PubMed: 1978555, related citations]

  182. Kwiatkowska, J., Kacprzak-Bergman, I. New erythrocyte glucose 6-phosphate dehydrogenase variant. Acta Haemat. 46: 188-192, 1971. [PubMed: 4256637, related citations] [Full Text]

  183. Kwok, C. J., Martin, A. C. R., Au, S. W. N., Lam, V. M. S. G6PDdb, an integrated database of glucose-6-phosphate dehydrogenase (G6PD) mutations. Hum. Mutat. 19: 217-224, 2002. [PubMed: 11857737, related citations] [Full Text]

  184. Lenzerini, L., Meera Khan, P., Filippi, G., Rattazzi, M. C., Rat, A. K. Characterization of glucose-6-phosphate dehydrogenase variants. I. Occurrence of a G6PD Seattle-like variant in Sardinia and its interaction with G6PD Mediterranean variant. Am. J. Hum. Genet. 21: 142-153, 1969. [PubMed: 5305539, related citations]

  185. Leopold, J. A., Dam, A., Maron, B. A., Scribner, A. W., Liao, R., Handy, D. E., Stanton, R. C., Pitt, B., Loscalzo, J. Aldosterone impairs vascular reactivity by decreasing glucose-6-phosphate dehydrogenase activity. Nature Med. 13: 189-197, 2007. Note: Erratum: Nature Med. 15: 1093 only, 2009. [PubMed: 17273168, images, related citations] [Full Text]

  186. Lisker, R., Linares, C., Motulsky, A. G. Glucose-6-phosphate dehydrogenase Mexico, a new variant with enzyme deficiency, abnormal mobility and absence of hemolysis. J. Lab. Clin. Med. 79: 788-793, 1972. [PubMed: 5018742, related citations]

  187. Lisker, R., Perez Briceno, R., Agrilar, L., Yoshida, A. A variant glucose-6-phosphate dehydrogenase Gd(-) Chiapas associated with moderate enzyme deficiency and occasional hemolytic anemia. Hum. Genet. 43: 81-84, 1978. [PubMed: 669721, related citations] [Full Text]

  188. Lisker, R., Perez Briceno, R., Zavala, C., Navarrette, J. I., Wessels, M., Yoshida, A. A glucose 6-phosphate dehydrogenase Gd(-) Castilla variant characterized by mild deficiency associated with drug-induced hemolytic anemia. J. Lab. Clin. Med. 90: 754-759, 1977. [PubMed: 903703, related citations]

  189. Lisker, R., Perez-Briceno, R., Beutler, E. A new glucose-6-phosphate dehydrogenase variant, Gd(-) Tepic, characterized by moderate enzyme deficiency and mild episodes of hemolytic anemia. Hum. Genet. 69: 19-21, 1985. [PubMed: 3967887, related citations] [Full Text]

  190. Lisker, R., Perez-Briceno, R., Rave, V., Yoshida, A. Glucosa-6-fosfato deshidrogenasa Gd(-) Distrito Federal: nueva variante asociada a deficiencia enzimatica moderada y anemia hemolitica ocasional. Rev. Invest. Clin. 33: 209-211, 1981. [PubMed: 7291768, related citations]

  191. Liu, Y., Phelan, J., Go, R. C. P., Prchal, J. F., Prchal, J. T. Rapid determination of clonality by detection of two closely-linked X chromosome exonic polymorphisms using allele-specific PCR. J. Clin. Invest. 99: 1984-1990, 1997. [PubMed: 9109443, related citations] [Full Text]

  192. Long, W. K., Kirkman, H. N., Sutton, H. H. Electrophoretically slow variants of glucose-6-phosphate dehydrogenase from red cells of Negroes. J. Lab. Clin. Med. 65: 81-87, 1965. [PubMed: 14260699, related citations]

  193. Longo, L., Vanegas, O. C., Patel, M., Rosti, V., Li, H., Waka, J., Merghoub, T., Pandolfi, P. P., Notaro, R., Manova, K., Luzzatto, L. Maternally transmitted severe glucose 6-phosphate dehydrogenase deficiency is an embryonic lethal. EMBO J. 21: 4229-4239, 2002. [PubMed: 12169625, images, related citations] [Full Text]

  194. Louicharoen, C., Nuchprayoon, I. G6PD Viangchan (871G-A) is the most common G6PD-deficient variant in the Cambodian population. J. Hum. Genet. 50: 448-452, 2005. [PubMed: 16155737, related citations] [Full Text]

  195. Louicharoen, C., Patin, E., Paul, R., Nuchprayoon, I., Witoonpanich, B., Peerapittayamongkol, C., Casademont, I., Sura, T., Laird, N. M., Singhasivanon, P., Quintana-Murci, L., Sakuntabhai, A. Positively selected G6PD-Mahidol mutation reduces Plasmodium vivax density in Southeast Asians. Science 326: 1546-1549, 2009. [PubMed: 20007901, related citations] [Full Text]

  196. Luzzatto, L., Afolayan, A. Enzyme properties of different types of human erythrocyte glucose-6-phosphate dehydrogenase with characterization of two new genetic variants. J. Clin. Invest. 47: 1833-1842, 1968. [PubMed: 5666113, related citations] [Full Text]

  197. Luzzatto, L., Usanga, E. A., Bienzle, U., Esan, G. F. J., Fusuan, F. A. Imbalance in X-chromosome expression: evidence for a human X-linked gene affecting growth of hemopoietic cells. Science 205: 1418-1420, 1979. [PubMed: 472761, related citations] [Full Text]

  198. Luzzatto, L., Usanga, E. A., Reddy, S. Glucose-6-phosphate dehydrogenase deficient red cells: resistance to infection by malarial parasites. Science 164: 839-842, 1969. [PubMed: 4889647, related citations] [Full Text]

  199. Luzzatto, L. Genetic heterogeneity and pathophysiology of G6PD deficiency. Brit. J. Haemat. 28: 151-156, 1974. [PubMed: 4154774, related citations] [Full Text]

  200. Luzzatto, L. Personal Communication. London, England 4/1990.

  201. MacDonald, D., Town, M., Mason, P., Vulliamy, T., Luzzatto, L., Goff, D. K. Deficiency in red blood cells. (Letter) Nature 350: 115, 1991. [PubMed: 2005960, related citations] [Full Text]

  202. Maeda, M., Constantoulakis, P., Chen, C.-S., Stamatoyannopoulos, G., Yoshida, A. Molecular abnormalities of a human glucose-6-phosphate dehydrogenase variant associated with undetectable enzyme activity and immunologically cross-reacting material. Am. J. Hum. Genet. 51: 386-395, 1992. [PubMed: 1353664, related citations]

  203. Mamlok, R. J., Mamlok, V., Mills, G. C., Daeschner, C. W., III, Schmalstieg, F. C., Anderson, D. C. Glucose-6-phosphate dehydrogenase deficiency, neutrophil dysfunction and Chromobacterium violaceum sepsis. J. Pediat. 111: 852-854, 1987. [PubMed: 3681551, related citations] [Full Text]

  204. Mamlok, R. J., Mills, G. C., Goldblum, R. M., Daeschner, C. W. Glucose-6-phosphate dehydrogenase Beaumont: a new variant with severe enzyme deficiency and chronic nonspherocytic hemolytic anemia. Enzyme 34: 15-21, 1985. [PubMed: 4085453, related citations] [Full Text]

  205. Mandelli, F., Amadori, S., DeLaurenzi, A., Kahn, A., Isacchi, G., Papa, G. Glucose-6-phosphate dehydrogenase Velletri: a new variant with reduced activity in a patient with congenital non-spherocytic haemolytic anemia. Acta Haemat. 57: 121-126, 1977. [PubMed: 14470, related citations] [Full Text]

  206. Marks, P. A., Banks, J., Gross, R. Genetic heterogeneity of glucose-6-phosphate dehydrogenase deficiency. Nature 194: 454-456, 1962.

  207. Martin, S. K., Miller, L. H., Alling, D., Okoye, V. C., Esan, G. J. F., Osunkoya, B. O., Deane, M. Severe malaria and glucose-6-phosphate-dehydrogenase deficiency: a reappraisal of the malaria-G6PD hypothesis. Lancet 313: 524-526, 1979. Note: Originally Volume I. [PubMed: 85108, related citations] [Full Text]

  208. Martini, G., Toniolo, D., Vulliamy, T., Luzzatto, L., Dono, R., Viglietto, G., Paonessa, G., D'Urso, M., Persico, M. G. Structural analysis of the X-linked gene encoding human glucose 6-phosphate dehydrogenase. EMBO J. 5: 1849-1855, 1986. [PubMed: 2428611, related citations] [Full Text]

  209. Mason, P. J., Stevens, D. J., Luzzatto, L., Brenner, S., Aparicio, S. Genomic structure and sequence of the Fugu rubripes glucose-6-phosphate dehydrogenase gene (G6PD). Genomics 26: 587-591, 1995. [PubMed: 7607684, related citations] [Full Text]

  210. Mason, P. J. New insights into G6PD deficiency. (Annotation) Brit. J. Haemat. 94: 585-591, 1996. [PubMed: 8826878, related citations]

  211. Matsuoka, H., Jichun, W., Hirai, M., Yoshida, S., Arai, M., Ishii, A., Baral, M. P. Two cases of glucose-6-phophate (sic) dehydrogenase-deficient Nepalese belonging to the G6PD Mediterranean-type, not India-Pakistan sub-type but Mediterranean-Middle East sub-type. J. Hum. Genet. 48: 275-277, 2003. [PubMed: 12768444, related citations] [Full Text]

  212. Matsuoka, H., Nguon, C., Kanbe, T., Jalloh, A., Sato, H., Yoshida, S., Hirai, M., Arai, M., Socheat, D., Kawamoto, F. Glucose-6-phosphate dehydrogenase (G6PD) mutations in Cambodia: G6PD Viangchan (871G-A) is the most common variant in the Cambodian population. J. Hum. Genet. 50: 468-472, 2005. [PubMed: 16136268, related citations] [Full Text]

  213. Matsuoka, H., Wang, J., Hirai, M., Arai, M., Yoshida, S., Kobayashi, T., Jalloh, A., Lin, K., Kawamoto, F. Glucose-6-phosphate dehydrogenase (G6PD) mutations in Myanmar: G6PD Mahidol (487G-A) is the most common variant in the Myanmar population. J. Hum. Genet. 49: 544-547, 2004. [PubMed: 15349799, related citations] [Full Text]

  214. McCann, S. R., Smithwick, A. M., Temperley, I. J., Tipton, K. G6PD (Dublin): chronic non-spherocytic haemolytic anaemia resulting from glucose-6-phosphate dehydrogenase deficiency in an Irish kindred. J. Med. Genet. 17: 191-193, 1980. [PubMed: 7401130, related citations] [Full Text]

  215. McCurdy, P. R., Blackwell, R. Q., Todd, D., Tso, S. C., Tuchinda, S. Further studies on glucose-6-phosphate dehydrogenase deficiency in Chinese subjects. J. Lab. Clin. Med. 75: 788-797, 1970. [PubMed: 5439677, related citations]

  216. McCurdy, P. R., Kamel, K., Selim, O. Heterogeneity of red cell glucose 6-phosphate dehydrogenase (G6PD) deficiency in Egypt. J. Lab. Clin. Med. 84: 673-680, 1974. [PubMed: 4283789, related citations]

  217. McCurdy, P. R., Kirkman, H. N., Naiman, J. L., Jim, R. T. S., Pickard, B. M. A Chinese variant of glucose-6-phosphate dehydrogenase. J. Lab. Clin. Med. 67: 374-385, 1966. [PubMed: 4379606, related citations]

  218. McCurdy, P. R., Mahmood, L. Red cell glucose-6-phosphate dehydrogenase deficiency in Pakistan. J. Lab. Clin. Med. 76: 943-948, 1970. [PubMed: 5485383, related citations]

  219. McCurdy, P. R., Maldonado, N. I., Dillon, D. E., Conrad, M. E. Variants of glucose-6-phosphate dehydrogenase (G-6-PD) associated with G-6-PD deficiency in Puerto Ricans. J. Lab. Clin. Med. 82: 432-437, 1973. [PubMed: 4728291, related citations]

  220. McCurdy, P. R. Use of genetic linkage for the detection of female carriers of hemophilia. New Eng. J. Med. 285: 218-219, 1971. [PubMed: 5087725, related citations] [Full Text]

  221. Meloni, T., Carta, F., Forteleoni, G., Carta, A., Ena, F., Meloni, G. F. Glucose 6-phosphate dehydrogenase deficiency and cataract of patients in northern Sardinia. Am. J. Ophthal. 110: 661-664, 1990. [PubMed: 2248331, related citations] [Full Text]

  222. Mentzer, W. C., Jr., Warner, R., Addiego, J., Smith, B., Walter, T. G6PD San Francisco: a new variant of glucose-6-phosphate dehydrogenase associated with congenital nonspherocytic hemolytic anemia. Blood 55: 195-198, 1980. [PubMed: 7353069, related citations]

  223. Mesbah-Namin, S. A., Sanati, M. H., Mowjoodi, A., Mason, P. J., Vulliamy, T. J., Noori-Daloii, M. R. Three major glucose-6-phosphate dehydrogenase-deficient polymorphic variants identified in Mazandaran state of Iran. Brit. J. Haemat. 117: 763-764, 2002. [PubMed: 12028056, related citations] [Full Text]

  224. Miller, D. R., Wollman, M. R. A new variant of glucose 6-phosphate dehydrogenase deficiency hereditary hemolytic anemia, G6PD Cornell: erythrocyte, leukocyte and platelet studies. Blood 44: 323-331, 1974. [PubMed: 4152748, related citations]

  225. Mills, G. C., Alperin, J. B., Trimmer, K. B. Studies on variant glucose-6-phosphate dehydrogenase: G6PD Fort Worth. Biochem. Med. 13: 264-275, 1975. [PubMed: 1007, related citations] [Full Text]

  226. Milner, G., Delamore, I. W., Yoshida, A. G-6-PD Manchester: a new variant associated with chronic nonspherocytic hemolytic anemia. Blood 43: 271-276, 1974. [PubMed: 4149233, related citations]

  227. Miwa, S., Fujii, H., Nakashima, K., Miura, Y., Yamada, K., Hagiwara, T., Fukuda, M. Three new electrophoretically normal glucose-6-phosphate dehydrogenase variants associated with congenital nonspherocytic hemolytic anemia found in Japan: G6PD Ogikubo, Yokohama, and Akita. Hum. Genet. 45: 11-17, 1978. [PubMed: 730178, related citations] [Full Text]

  228. Miwa, S., Fujii, H., Nakatsuji, T., Ishida, Y., Oda, E., Kaneto, A., Motokawa, M., Ariga, Y., Fukuchi, S., Sasai, S., Hiraoka, K., Kashii, H., Kodama, T. Four new electrophoretically slow-moving glucose-6-phosphate dehydrogenase variants associated with congenital nonspherocytic hemolytic anemia found in Japan: Gd(-) Kurume, Gd(-) Fukushima, Gd(-) Yamaguchi, and Gd(-) Wakayama. Am. J. Hemat. 5: 131-138, 1978. [PubMed: 736032, related citations] [Full Text]

  229. Miwa, S., Fujii, H. Molecular basis of erythroenzymopathies associated with hereditary hemolytic anemia: tabulation of mutant enzymes. Am. J. Hemat. 51: 122-132, 1996. [PubMed: 8579052, related citations] [Full Text]

  230. Miwa, S., Nakashima, K., Ono, J., Fujii, H., Suzuki, E. Three glucose 6-phosphate dehydrogenase variants found in Japan. Hum. Genet. 36: 327-334, 1977. [PubMed: 15940, related citations] [Full Text]

  231. Miwa, S., Ono, J., Nakashima, K., Abe, S., Kageoka, T., Shinohara, K., Isobe, J., Yamaguchi, H. Two new glucose 6-phosphate dehydrogenase variants associated with congenital nonspherocytic hemolytic anemia found in Japan: Gd(-) Tokushima and Gd(-) Tokyo. Am. J. Hemat. 1: 433-442, 1976. [PubMed: 1008056, related citations] [Full Text]

  232. Modiano, G., Battistuzzi, G., Esan, G. J. F., Testa, U., Luzzatto, L. Genetic heterogeneity of 'normal' human erythrocyte glucose-6-phosphate dehydrogenase: an isoelectrophoretic polymorphism. Proc. Nat. Acad. Sci. 76: 852-856, 1979. [PubMed: 284409, related citations] [Full Text]

  233. Mohrenweiser, H. W., Neel, J. V. Frequency of thermostability variants: estimation of total 'rare' variant frequency in human populations. Proc. Nat. Acad. Sci. 78: 5729-5733, 1981. [PubMed: 6946512, related citations] [Full Text]

  234. Morelli, A., Benatti, U., Guida, L., De Flora, A. G6PD Cagliari: a new low activity glucose 6-phosphate dehydrogenase variant characterized by enhanced intracellular lability. Hum. Genet. 66: 62-65, 1984. [PubMed: 6698555, related citations] [Full Text]

  235. Morisaki, T., Fujii, H., Takegawa, S., Tani, K., Hirono, A., Takizawa, T., Takahashi, K., Shinogi, M., Teshirogi, T., Miwa, S. G6PD Sendagi: a new glucose-6-phosphate dehydrogenase variant associated with congenital hemolytic anemia. Hum. Genet. 65: 214-215, 1983. [PubMed: 6654337, related citations] [Full Text]

  236. Nafa, K., Reghis, A., Osmani, N., Baghli, L., Benabadji, M., Kaplan, J.-C., Vulliamy, T. J., Luzzatto, L. G6PD Aures: a new mutation (48 ile-to-thr) causing mild G6PD deficiency is associated with favism. Hum. Molec. Genet. 2: 81-82, 1993. [PubMed: 8490627, related citations] [Full Text]

  237. Nagel, R. L., Ranney, H. M. Genetic epidemiology of structural mutations of the beta-globin gene. Semin. Hemat. 27: 342-359, 1990. [PubMed: 2255919, related citations]

  238. Nakai, T., Yoshida, A. G6PD Heian. A glucose-6-phosphate dehydrogenase variant associated with hemolytic anemia found in Japan. Clin. Chim. Acta 51: 199-203, 1974. [PubMed: 4828221, related citations] [Full Text]

  239. Nakashima, K., Ono, J., Abe, S., Miwa, S., Yoshida, A. G6PD Ube: a glucose 6-phosphate dehydrogenase variant found in four unrelated Japanese families. Am. J. Hum. Genet. 29: 24-30, 1977. [PubMed: 835572, related citations]

  240. Nakatsuji, T., Miwa, S. Incidence and characteristics of glucose-6-phosphate dehydrogenase variants in Japan. Hum. Genet. 51: 297-305, 1979. [PubMed: 511159, related citations] [Full Text]

  241. Nance, W. E. Turner's syndrome, twinning, and an unusual variant of glucose-6-phosphate dehydrogenase. Am. J. Hum. Genet. 16: 380-392, 1964. [PubMed: 14207553, related citations]

  242. Necheles, T. F., Snyder, L. M., Strauss, W. Glucose-6-phosphate dehydrogenase Boston. A new variant associated with congenital nonspherocytic hemolytic disease. Humangenetik 13: 218-221, 1971. [PubMed: 4398877, related citations] [Full Text]

  243. Niazi, G., Adeyokunu, A., Westwood, B., Beutler, E. G6PD Aures: a rare mutant of G6PD in Saudi Arabia: molecular and clinical presentations. Saudi Med. J. 17: 311-314, 1996.

  244. Ninfali, P., Baronciani, L., Bardoni, A., Bresolin, N. Muscle expression of glucose-6-phosphate dehydrogenase deficiency in different variants. Clin. Genet. 48: 232-237, 1995. [PubMed: 8825599, related citations] [Full Text]

  245. Ninokata, A., Kimura, R., Samakkarn, U., Settheetham-Ishida, W., Ishida, T. Coexistence of five G6PD variants indicates ethnic complexity of Phuket islanders, Southern Thailand. J. Hum. Genet. 51: 424-428, 2006. [PubMed: 16528451, related citations] [Full Text]

  246. Notaro, R., Afolayan, A., Luzzatto, L. Human mutations in glucose 6-phosphate dehydrogenase reflect evolutionary history. FASEB J. 14: 485-494, 2000. [PubMed: 10698963, related citations] [Full Text]

  247. Nowicki, L., Strobel, S., Martin, H., Koschwitz, U. Ueber eine neue erythrocytaere glucose 6-phosphatdehydrogenase Variante, Typ Frankfurt. Klin. Wschr. 52: 478-484, 1974. [PubMed: 4408431, related citations] [Full Text]

  248. Nsouly, G. M., Prchal, J. T. Characterization of a new G6PD variant and its associated oxidative damage. Clin. Res. 29: 829, 1981.

  249. O'Brien, S. J. The extent and character of biochemical genetic variation in the domestic cat. J. Hered. 71: 2-8, 1980.

  250. Ogura, H., Morisaki, T., Tani, K., Kanno, H., Tsutsumi, H., Takahashi, K., Miyamori, T., Fujii, H., Miwa, S. A new glucose-6-phosphate dehydrogenase variant (G6PD Tsukui) associated with congenital hemolytic anemia. Hum. Genet. 78: 369-371, 1988. [PubMed: 3360447, related citations] [Full Text]

  251. Ohno, S. Sex Chromosomes and Sex-linked Genes. Berlin and New York: Springer (pub.) 1967.

  252. Othieno-Obel, A. East African variant of glucose-6-phosphate dehydrogenase. East Afr. Med. J. 49: 230-234, 1972. [PubMed: 5047808, related citations]

  253. Pai, G. S., Sprenkle, J. A., Do, T. T., Mareni, C. E., Migeon, B. R. Localization of loci for hypoxanthine phosphoribosyltransferase and glucose-6-phosphate dehydrogenase and biochemical evidence of nonrandom X chromosome expression from studies of a human X-autosome translocation. Proc. Nat. Acad. Sci. 77: 2810-2813, 1980. [PubMed: 6930669, related citations] [Full Text]

  254. Panich, V., Bumrungtrakul, P., Jitjai, C., Kamolmatayakul, S., Khoprasert, B., Klaisuvan, C., Kongmuang, U., Maneechai, P., Pornpatkul, M., Ruengrairatanaroje, P., Surapruk, P., Viriyayudhakorn, S. Glucose-6-phosphate dehydrogenase deficiency in South Vietnamese. Hum. Hered. 30: 361-364, 1980. [PubMed: 7216225, related citations] [Full Text]

  255. Panich, V., Na-Nakorn, S. G6PD variants in Thailand. J. Med. Assoc. Thai. 63: 537-543, 1980. [PubMed: 7441068, related citations]

  256. Panich, V., Sungnate, T., Na-Nakorn, S. Acute intravascular hemolysis and renal failure in a new glucose 6-phosphate dehydrogenase variant: G6PD Siriraj. J. Med. Assoc. Thai. 55: 726-731, 1972. [PubMed: 4650733, related citations]

  257. Panich, V., Sungnate, T., Wasi, P., Na-Nakorn, S. G-6-PD Mahidol: the most common glucose-6-phosphate dehydrogenase variant in Thailand. J. Med. Assoc. Thai. 55: 576-585, 1972. [PubMed: 5081671, related citations]

  258. Panich, V., Sungnate, T. Characterization of glucose 6-phosphate dehydrogenase in Thailand: the occurrence of 6 variants among 50 G6PD deficient Thai. Humangenetik 18: 39-46, 1973. [PubMed: 4721339, related citations]

  259. Panich, V. G6PD characterization in Thailand. Genetics 74 (suppl.): s208 only, 1973.

  260. Panich, V. G6PD Intanon, a new glucose 6-phosphate dehydrogenase variant. Humangenetik 21: 203-205, 1974. [PubMed: 4837298, related citations] [Full Text]

  261. Panich, V. G6PD variants in Laotians. Hum. Hered. 24: 285-290, 1974. [PubMed: 4435794, related citations] [Full Text]

  262. Panich, V. Glucose-6-phosphate dehydrogenase in Thailand. Hum. Genet. 53: 227-228, 1980. [PubMed: 7358391, related citations] [Full Text]

  263. Pawlak, A. L., Mazurkiewicz, C. A., Ordynski, J., Ruzynkowa, D., Horst, A. G6PD Poznan, variant with severe enzyme deficiency. Humangenetik 28: 163-165, 1975. [PubMed: 1150271, related citations] [Full Text]

  264. Pawlak, A. L., Zagorski, Z., Rozynkowa, D., Horst, A. Polish variant of glucose-6-phosphate dehydrogenase (G-6-PD Lublin). Humangenetik 10: 340-343, 1970. [PubMed: 5493238, related citations] [Full Text]

  265. Pekrun, A., Eber, S. W., Schroter, W. G6PD Avenches and G6PD Moosburg: biochemical and erythrocyte membrane characterization. Blut 58: 11-14, 1989. [PubMed: 2917200, related citations] [Full Text]

  266. Perroni, L., Tassara, P., Baldi, M., Reali, R., Scartezzini, P. G6PD variants detected in Genoa area. In: Weatherall, D. J.; Fiorelli, G.; Gorini, S.: Advances in Red Blood Cell Biology. New York: Raven Press (pub.) 1982. Pp. 409-416.

  267. Persico, M. G., Toniolo, D., Nobile, C., D'Urso, M., Luzzatto, L. cDNA sequences of human glucose 6-phosphate dehydrogenase cloned in pBR322. Nature 294: 778-780, 1981. [PubMed: 6895658, related citations] [Full Text]

  268. Persico, M. G., Viglietto, G., Martini, G., Toniolo, D., Paonessa, G., Moscatelli, C., Dono, R., Vulliamy, T., Luzzatto, L., D'Urso, M. Isolation of human glucose-6-phosphate dehydrogenase (G6PD) cDNA clones: primary structure of the protein and unusual 5-prime non-coding region. Nucleic Acids Res. 14: 2511-2522, 1986. Note: Erratum: Nucleic Acids Res. 14: 7822 only, 1986. [PubMed: 3515319, related citations] [Full Text]

  269. Picat, C., Etiemble, J., Boivin, P., Le Prise, P.-Y. Gd(-) Rennes, a new deficient variant of glucose-6-phosphate dehydrogenase associated with congenital nonspherocytic hemolytic anemia found in France. Hum. Genet. 55: 125-127, 1980. [PubMed: 7450748, related citations] [Full Text]

  270. Pinto, P. V. C., Newton, W. A., Jr., Richardson, K. E. Evidence for four types of erythrocyte glucose-6-phosphate dehydrogenase from G-6-PD deficient human subjects. J. Clin. Invest. 45: 823-831, 1966. [PubMed: 5913292, related citations] [Full Text]

  271. Poggi, V. Personal Communication. London, England 1989.

  272. Poon, M.-C., Hall, K., Scott, C. W., Prchal, J. T. G6PD Viangchan: a new glucose 6-phosphate dehydrogenase variant from Laos. Hum. Genet. 78: 98-99, 1988. [PubMed: 3338798, related citations] [Full Text]

  273. Porter, I. H., Boyer, S. H., Watson-Williams, E. J., Adam, A., Szeinberg, A., Siniscalco, M. Variation of glucose-6-phosphate dehydrogenase in different populations. Lancet 283: 895-899, 1964. Note: Originally Volume I. [PubMed: 14124079, related citations] [Full Text]

  274. Porter, I. H., Schulze, J., McKusick, V. A. Genetical linkage between the loci for glucose-6-phosphate dehydrogenase deficiency and colour-blindness in American Negroes. Ann. Hum. Genet. 26: 107-122, 1962. [PubMed: 13985567, related citations] [Full Text]

  275. Prchal, J., Moreno, H., Conrad, M., Vitek, A. G-6-PD Dothan: a new variant associated with chronic hemolytic anemia. I.R.C.S. 7: 348, 1979.

  276. Prchal, J. T., Crist, W. M., Malluh, A., Vitek, A., Tauxe, W. N., Carroll, A. J. A new glucose-6-phosphate dehydrogenase deficient variant in a patient with Chediak-Higashi syndrome. Blood 56: 476-480, 1980. [PubMed: 7407414, related citations]

  277. Prchal, J. T., Hall, K., Csepreghy, M., Lilly, M., Berkow, R., Scott, C. W. Two apparent glucose-6-phosphate dehydrogenase variants in normal XY males: G6PD Alabama. Am. J. Med. 84: 517-523, 1988. [PubMed: 3348252, related citations] [Full Text]

  278. Ramot, B., Ben-Bassat, I., Shchory, M. New glucose-6-phosphate dehydrogenase variants observed in Israel and their association with congenital nonspherocytic hemolytic disease. J. Lab. Clin. Med. 74: 895-901, 1969. [PubMed: 5359663, related citations]

  279. Ramot, B., Brok, F. A new glucose-6-phosphate dehydrogenase mutant (Tel-Hashomer mutant). Ann. Hum. Genet. 28: 167-172, 1964. [PubMed: 14228003, related citations] [Full Text]

  280. Rattazzi, M. C., Corash, L. M., Van Zzanen, G. E., Jaffe, E. R., Piomelli, S. G6PD deficiency and chronic hemolysis: four new mutants--relationships between clinical syndrome and enzyme kinetics. Blood 38: 205-218, 1971. [PubMed: 4397482, related citations]

  281. Rattazzi, M. C., Lenzerini, L., Meera Khan, P., Luzzatto, L. Characterization of glucose-6-phosphate dehydrogenase variants. II. G6PD Kephalonia, G6PD Attica, and G6PD 'Seattle-like' found in Greece. Am. J. Hum. Genet. 21: 154-167, 1969. [PubMed: 5770172, related citations]

  282. Ravindranath, Y., Beutler, E. Two new variants of glucose-6-phosphate dehydrogenase associated with hereditary non-spherocytic hemolytic anemia: G6PD Wayne and G6PD Huron. Am. J. Hemat. 24: 357-363, 1987. [PubMed: 3565372, related citations] [Full Text]

  283. Reys, L., Manso, C., Stamatoyannopoulos, G. Genetic studies on Southeastern Bantu of Mozambique. I. Variants of glucose-6-phosphate dehydrogenase. Am. J. Hum. Genet. 22: 203-215, 1970. [PubMed: 5435642, related citations]

  284. Rosenstraus, M., Chasin, L. A. Isolation of mammalian cell mutants deficient in glucose-6-phosphate dehydrogenase activity: linkage to hypoxanthine phosphoribosyl transferase. Proc. Nat. Acad. Sci. 72: 493-497, 1975. [PubMed: 1054832, related citations] [Full Text]

  285. Roth, E. F., Jr., Raventos-Suarez, C., Rinaldi, A., Nagel, R. L. Glucose-6-phosphate dehydrogenase deficiency inhibits in vitro growth of Plasmodium falciparum. Proc. Nat. Acad. Sci. 80: 298-299, 1983. [PubMed: 6337374, related citations] [Full Text]

  286. Roychoudhury, A. K., Nei, M. Human Polymorphic Genes: World Distribution. New York: Oxford Univ. Press (pub.) 1988.

  287. Ruwende, C., Khoo, S. C., Snow, R. W., Yates, S. N. R., Kwiatkoweld, D., Gupta, S., Warn, P., Alisopp, G. E. M., Gilbert, S. C., Peschu, N., Newbold, C. I., Greenwood, S. M., Marsh, K., Hill, A. V. S. Natural selection of hemi- and heterozygotes for G6PD deficiency in Africa by resistance to severe malaria. Nature 376: 246-249, 1995. [PubMed: 7617034, related citations] [Full Text]

  288. Saenz, G. F., Chaves, M., Berrantes, A., Elizondo, J., Montero, A. G., Yoshida, A. A glucose-6-phosphate dehydrogenase variant, Gd(-) Santamaria found in Costa Rica. Acta Haemat. 72: 37-40, 1984. [PubMed: 6433630, related citations] [Full Text]

  289. Saidi, N., Hors-Cayla, M. C., Van Cong, N., Benne, F. Sheep gene mapping by somatic cell hybridization. (Abstract) Cytogenet. Cell Genet. 25: 200, 1979.

  290. Samuel, A. P. W., Saha, N., Omer, A., Hoffbrand, A. V. Quantitative expression of G6PD activity of different phenotypes of G6PD and haemoglobin in a Sudanese population. Hum. Hered. 31: 110-115, 1981. [PubMed: 7228057, related citations] [Full Text]

  291. Sansone, G., Perroni, L., Testa, U., Mareni, C., Luzzatto, L. New genetic variants of glucose 6-phosphate dehydrogenase (G6PD) in Italy. Ann. Hum. Genet. 45: 97-104, 1981. [PubMed: 7316485, related citations] [Full Text]

  292. Sansone, G., Perroni, L., Yoshida, A., Dave, V. A new glucose 6-phosphate dehydrogenase variant (Gd Trinacria) in two unrelated families of Sicilian ancestry. Ital. J. Biochem. 26: 44-50, 1977. [PubMed: 16832, related citations]

  293. Sansone, G., Perroni, L., Yoshida, A. Glucose-6-phosphate dehydrogenase variants from Italian subjects associated with severe neonatal jaundice. Brit. J. Haemat. 31: 159-165, 1975. [PubMed: 1201235, related citations] [Full Text]

  294. Shatskaya, T. L., Krasnopolskaya, K. D., Annenkov, G. A. A description of new mutant forms of erythrocyte glucose-6-phosphate dehydrogenase isolated at the territory of the Soviet Union. Genetika 11: 116-122, 1975.

  295. Shatskaya, T. L., Krasnopolskaya, K. D., Idelson, L. J. Mutant forms of erythrocyte glucose 6-phosphate dehydrogenase in Ashkenazi: description of two new variants, G6PD Kirovograd and G6PD Zhitomir. Hum. Genet. 33: 175-178, 1976. [PubMed: 133080, related citations] [Full Text]

  296. Shatskaya, T. L., Krasnopolskaya, K. D., Idelson, L. J. The new form of glucose 6-phosphate dehydrogenase (G6PD 'Kaluga') from erythrocytes of a patient with chronic non-spherocytic hemolytic anemia. Vopr. Med. Khim. 22: 764-768, 1976. [PubMed: 17221, related citations]

  297. Shatskaya, T. L., Krasnopolskaya, K. D., Tzoneva, M., Mavrudieva, M., Toncheva, D. Variants of erythrocyte glucose-6-phosphate dehydrogenase (G6PD) in Bulgarian populations. Hum. Genet. 54: 115-117, 1980. [PubMed: 7390473, related citations] [Full Text]

  298. Shatskaya, T. L., Krasnopolskaya, K. D., Zakharova, T. V. Regularities of distribution of Gd-alleles in Azerbaijan. II. Identification of G6PD mutant forms. Genetika 16: 2217-2225, 1980.

  299. Shows, T. B., Brown, J. A., Chapman, V. M. Comparative gene mapping of HPRT, G6PD and PGK in man, mouse, and Muntjac deer. Birth Defects Orig. Art. Ser. XII(7): 436-439, 1976. [PubMed: 1024651, related citations]

  300. Shows, T. B., Brown, J. A. Human X-linked genes regionally mapped utilizing X-autosome translocations and somatic cell hybrids. Proc. Nat. Acad. Sci. 72: 2125-2129, 1975. [PubMed: 1056018, related citations] [Full Text]

  301. Shows, T. B., Tashian, R. E., Brewer, G. J. Erythrocyte glucose-6-phosphate dehydrogenase in Caucasians: new inherited variant. Science 145: 1056-1057, 1964. [PubMed: 14172618, related citations] [Full Text]

  302. Sidi, Y., Aderka, D., Brok-Simoni, F., Benjamin, D., Ramot, B., Pinkhas, J. Viral hepatitis with extreme hyperbilirubinemia, massive hemolysis and encephalopathy in a patient with a new G6PD variant. Isr. J. Med. Sci. 16: 130-133, 1980. [PubMed: 7364565, related citations]

  303. Siegel, N. H., Beutler, E. Hemolytic anemia caused by G-6-PD Carswell, a new variant. Ann. Intern. Med. 75: 437-439, 1971. [PubMed: 5568155, related citations] [Full Text]

  304. Smith, J. E., Ryer, K., Wallace, L. Glucose-6-phosphate dehydrogenase deficiency in a dog. Enzyme 21: 379-382, 1976. [PubMed: 939229, related citations] [Full Text]

  305. Snyder, L. M., Necheles, T. F., Reddy, W. J. G-6-PD Worcester: a new variant, associated with X-linked optic atrophy. Am. J. Med. 49: 125-132, 1970. [PubMed: 5311120, related citations] [Full Text]

  306. Stamatoyannopoulos, G., Kotsakis, P., Voigtlander, V., Akrivakis, A., Motulsky, A. G. Electrophoretic diversity of glucose-6-phosphate dehydrogenase among Greeks. Am. J. Hum. Genet. 22: 587-596, 1970. [PubMed: 5516240, related citations]

  307. Stamatoyannopoulos, G., Voigtlander, V., Akrivakis, A. Thessaly variant of glucose-6-phosphate dehydrogenase. Humangenetik 9: 23-25, 1970. [PubMed: 5496124, related citations] [Full Text]

  308. Stamatoyannopoulos, G., Voigtlander, V., Kotsakis, P., Akrivakis, A. Genetic diversity of the 'Mediterranean' glucose-6-phosphate dehydrogenase deficiency phenotype. J. Clin. Invest. 50: 1253-1261, 1971. [PubMed: 4397065, related citations] [Full Text]

  309. Stamatoyannopoulos, G., Yoshida, A., Bacopoulos, C., Motulsky, A. G. Athens variant of glucose-6-phosphate dehydrogenase. Science 157: 831-833, 1967. [PubMed: 17842795, related citations] [Full Text]

  310. Stevens, D. J., Wanachiwanawin, W., Mason, P. J., Vulliamy, T. J., Luzzatto, L. G6PD Canton a common deficient variant in South East Asia caused by a 459 arg-to-leu mutation. Nucleic Acids Res. 18: 7190, 1990. [PubMed: 2263506, related citations] [Full Text]

  311. Stocco dos Santos, R. C., Barretto, O. C. O., Nonoyama, K., Castro, N. H. C., Ferraz, O. P., Walter-Moura, J., Vescio, C. C. S., Becak, W. X-linked syndrome: mental retardation, hip luxation, and G6PD variant (Gd(+) Butantan). Am. J. Med. Genet. 39: 133-136, 1991. [PubMed: 2063914, related citations] [Full Text]

  312. Stocco dos Santos, R. C., Castro, N. H. C., Holmes, A. L., Becak, W., Tackels-Horne, D., Lindsey, C. J., Lubs, H. A., Stevenson, R. E., Schwartz, C. E. Stocco dos Santos X-linked mental retardation syndrome: clinical elucidation and localization to Xp11.3-Xq21.3. Am. J. Med. Genet. 118A: 255-259, 2003. [PubMed: 12673656, related citations] [Full Text]

  313. Streiff, F., Vigneron, C. Anemie hemolytique chronique par deficit en glucose 6-phosphate deshydrogenase dans une famille d'origine Lorraine. Nouv. Rev. Franc. Hemat. 11: 279-290, 1971. [PubMed: 4252680, related citations]

  314. Takahashi, K., Fujii, H., Takegawa, S., Tani, K., Hirono, A., Takizawa, T., Kawakatsu, T., Miwa, S. A new glucose-6-phosphate dehydrogenase variant (G6PD Nagano) associated with congenital hemolytic anemia. Hum. Genet. 62: 368-370, 1982. [PubMed: 7166314, related citations] [Full Text]

  315. Takizawa, T., Fujii, H., Takegawa, S., Takahashi, K., Hirono, A., Morisaki, T., Kanno, H., Oka, R., Yoshioka, H., Miwa, S. A unique electrophoretic slow-moving glucose 6-phosphate dehydrogenase variant (G6PD Asahikawa) with a markedly acidic pH optimum. Hum. Genet. 68: 70-72, 1984. [PubMed: 6500558, related citations] [Full Text]

  316. Takizawa, T., Huang, I.-Y., Ikuta, T., Yoshida, A. Human glucose-6-phosphate dehydrogenase: primary structure and cDNA cloning. Proc. Nat. Acad. Sci. 83: 4157-4161, 1986. [PubMed: 3012556, related citations] [Full Text]

  317. Takizawa, T., Yoneyama, Y., Miwa, S., Yoshida, A. A single nucleotide base transition is the basis of the common human glucose-6-phosphate dehydrogenase variant A(+). Genomics 1: 228-231, 1987. [PubMed: 3446582, related citations] [Full Text]

  318. Takizawa, T., Yoshida, A. Molecular abnormality of the common glucose-6-phosphate dehydrogenase variant, G6PD A(+), and restriction-fragment-length polymorphism. (Abstract) Am. J. Hum. Genet. 41: A241, 1987.

  319. Talalak, P., Beutler, E. G-6-PD Bangkok: a new variant found in congenital nonspherocytic hemolytic disease (CNHD). Blood 33: 772-776, 1969. [PubMed: 5779160, related citations]

  320. Tanaka, K. R., Beutler, E. Hereditary hemolytic anemia due to glucose-6-phosphate dehydrogenase Torrance: a new variant. J. Lab. Clin. Med. 73: 657-667, 1969. [PubMed: 4388268, related citations]

  321. Tang, T. K., Huang, C.-S., Huang, M. J., Tam, K.-B., Yeh, C.-H., Tang, C.-J. C. Diverse point mutations result in glucose-6-phosphate dehydrogenase (G6PD) polymorphism in Taiwan. Blood 79: 2135-2140, 1992. [PubMed: 1562739, related citations]

  322. Testa, U., Meloni, T., Lania, A., Battistuzzi, G., Cutillo, S., Luzzatto, L. Genetic heterogeneity of glucose-6-phosphate dehydrogenase deficiency in Sardinia. Hum. Genet. 56: 99-105, 1980. [PubMed: 7203486, related citations] [Full Text]

  323. Thigpen, J. T., Steinberg, M. H., Beutler, E., Gillespie, G. T., Jr., Dreiling, B. J., Morrison, F. S. Glucose-6-phosphate dehydrogenase Jackson, a new variant associated with hemolytic anemia. Acta Haemat. 51: 310-314, 1974. [PubMed: 4209318, related citations] [Full Text]

  324. Tishkoff, S. A., Varkonyi, R., Cahinhinan, N., Abbes, S., Argyropoulos, G., Destro-Bisol, G., Drousiotou, A., Dangerfield, B., Lefranc, G., Loiselet, J., Piro, A., Stoneking, M., Tagarelli, A., Tagarelli, G., Touma, E. H., Williams, S. M., Clark, A. G. Haplotype diversity and linkage disequilibrium at human G6PD: recent origin of alleles that confer malarial resistance. Science 293: 455-462, 2001. [PubMed: 11423617, related citations] [Full Text]

  325. Tokarev, Y. N., Chernyak, N. B., Batischev, A. I., Lanzina, N. V., Alexeyev, G. A. Etude des proprietes electrophoretiques et cinetiques de la glucose-6-phosphate deshydrogenase (Gd) d'erythrocytes dans les deficits hereditaires de l'enzyme: description d'une nouvelle variante de glucose-6-phosphate deshydrogenase: la Gd Kremenchug. Nouv. Rev. Franc. Hemat. 20: 557-564, 1978. [PubMed: 752149, related citations]

  326. Toncheva, D., Tzoneva, M. Genetic polymorphism of G6PD in a Bulgarian population. Hum. Genet. 67: 340-342, 1984. [PubMed: 6540752, related citations] [Full Text]

  327. Toncheva, D. Variants of glucose-6-phosphate dehydrogenase in a Vietnamese population. Hum. Hered. 36: 348-351, 1986. [PubMed: 3793116, related citations] [Full Text]

  328. Toniolo, D., Martini, G., Migeon, B. R., Dono, R. Expression of the G6PD locus on the human X chromosome is associated with demethylation of three CpG islands within 100 kb of DNA. EMBO J. 7: 401-406, 1988. [PubMed: 2835227, related citations] [Full Text]

  329. Town, M., Athanasiou-Metaxa, M., Luzzatto, L. Intragenic interspecific complementation of glucose 6-phosphate dehydrogenase in human-hamster cell hybrids. Somat. Cell Molec. Genet. 16: 97-108, 1990. [PubMed: 2157298, related citations] [Full Text]

  330. Town, M., Bautista, J. M., Mason, P. J., Luzzatto, L. Both mutations in G6PD A- are necessary to produce the G6PD deficient phenotype. Hum. Molec. Genet. 1: 171-174, 1992. [PubMed: 1303173, related citations] [Full Text]

  331. Usanga, E. A., Bienzle, U., Cancedda, K., Fasuan, F. A., Ajayi, O., Luzzatto, L. Genetic variants of human erythrocyte glucose 6-phosphate dehydrogenase: new variants in West Africa characterized by column chromatography. Ann. Hum. Genet. 40: 279-286, 1977. [PubMed: 848857, related citations] [Full Text]

  332. Vaca, G., Ibarra, B., Garcia Cruz, D., Medina, C., Romero, F., Cantu, J. M., Beutler, E. G-6-PD Jalisco and G-6-PD Morelia: two new Mexican variants. Hum. Genet. 71: 82-85, 1985. [PubMed: 4029955, related citations] [Full Text]

  333. Vaca, G., Ibarra, B., Romero, F., Olivares, N., Cantu, J. M., Beutler, E. G-6-PD Guadalajara: a new mutant associated with chronic nonspherocytic hemolytic anemia. Hum. Genet. 61: 175-176, 1982. [PubMed: 7129446, related citations] [Full Text]

  334. van Bruggen, R., Bautista, J. M., Petropoulou, T., de Boer, M., van Zwieten, R., Gomez-Gallego, F., Belohradsky, B. H., Hartwig, N. G., Stevens, D., Mason, P. J., Roos, D. Deletion of leucine 61 in glucose-6-phosphate dehydrogenase leads to chronic nonspherocytic anemia, granulocyte dysfunction, and increased susceptibility to infections. Blood 100: 1026-1030, 2002. [PubMed: 12130518, related citations] [Full Text]

  335. Ventura, A., Panizon, F., Soranzo, M. R., Veneziano, G., Sansone, G., Testa, U., Luzzatto, L. Congenital dyserythropoietic anaemia type II associated with a new type of G6PD deficiency (G6PD Gabrovizza). Acta Haemat. 71: 227-234, 1984. [PubMed: 6426235, related citations] [Full Text]

  336. Vergnes, H., Gherardi, M., Bouloux, C. Erythrocyte glucose-6-phosphate dehydrogenase in the Niokolonko (Malinke of the Niokolo) of the Eastern Senegal: identification of a slow variant with normal activity (Tacoma-like). Hum. Hered. 25: 80-87, 1975. [PubMed: 1150303, related citations] [Full Text]

  337. Vergnes, H., Gherardi, M., Quilici, J. C., Yoshida, A., Giacardy, R. G6PD Luz-Saint-Sauveur: a new variant with abnormal electrophoretic mobility, mild enzyme deficiency and absence of hemolytic disorders. I.R.C.S. 7: 14, 1973.

  338. Vergnes, H., Gherardi, M., Yoshida, A. G6PD Lozere and Trinacria-like: segregation of two non-hemolytic variants in a French family. Hum. Genet. 34: 293-298, 1976. [PubMed: 1002152, related citations] [Full Text]

  339. Vergnes, H., Ribet, A., Bommelaer, G., Amadieu, J., Brun, H. GD(-) Muret and GD(-) Colomiers, two new variants of glucose-6-phosphate dehydrogenase associated with favism. Hum. Genet. 57: 332-334, 1981. [PubMed: 7250973, related citations] [Full Text]

  340. Vergnes, H., Yoshida, A., Gourdin, D., Gherardi, M., Bierme, R., Ruffie, J. Glucose 6-phosphate dehydrogenase Toulouse: a new variant with marked instability and severe deficiency discovered in a family of Mediterranean ancestry. Acta Haemat. 51: 240-249, 1974. [PubMed: 4152602, related citations] [Full Text]

  341. Viglietto, G., Montanaro, V., Calabro, V., Vallone, D., D'Urso, M., Persico, M. G., Battistuzzi, G. Common glucose-6-phosphate dehydrogenase (G6PD) variants from the Italian population: biochemical and molecular characterization. Ann. Hum. Genet. 54: 1-15, 1990. [PubMed: 2321910, related citations] [Full Text]

  342. Vives-Corrons, J. L., Feliu, E., Pujades, M. A., Cardellach, F., Rozman, C., Carreras, A., Jou, J. M., Vallespi, M. T., Zuazu, F. J. Severe glucose-6-phosphate dehydrogenase (G6PD) deficiency associated with chronic hemolytic anemia, granulocyte dysfunction, and increased susceptibility to infections: description of a new molecular variant (G6PD Barcelona). Blood 59: 428-434, 1982. [PubMed: 7055648, related citations]

  343. Vives-Corrons, J. L., Pujades, A., Curia, M. D. Caracterizacion molecular de la glucosa-6-fosfato deshidrogenasa (G6PD) en 24 casos de deficit enzimatico y descripcion de una nueva variante (G6PD-Betica). Sangre 25: 1049-1064, 1980. [PubMed: 7221827, related citations]

  344. Vives-Corrons, J. L., Pujades, A. Heterogeneity of 'Mediterranean type' glucose-6-phosphate dehydrogenase (G6PD) deficiency in Spain and description of two new variants associated with favism. Hum. Genet. 60: 216-221, 1982. [PubMed: 7106752, related citations] [Full Text]

  345. Vives-Corrons, J. L., Pujades, M. A., Petit, J., Colomer, D., Corbella, M., Aguilar i Bascompte, J. L., Merino, A. Chronic nonspherocytic hemolytic anemia (CNSHA) and glucose 6 phosphate dehydrogenase (G6PD) deficiency in a patient with familial amyloidotic polyneuropathy (FAP): molecular study of a new variant (G6PD Clinic) with markedly acidic pH optimum. Hum. Genet. 81: 161-164, 1989. [PubMed: 2912886, related citations] [Full Text]

  346. Vives-Corrons, J.-L., Kuhl, W., Pujades, M. A., Beutler, E. Molecular genetics of the glucose-6-phosphate dehydrogenase (G6PD) Mediterranean variant and description of a new G6PD mutant, G6PD Andalus(1361A). Am. J. Hum. Genet. 47: 575-579, 1990. [PubMed: 2393028, related citations]

  347. Vulliamy, T., Beutler, E., Luzzatto, L. Variants of glucose-6-phosphate dehydrogenase are due to missense mutations spread throughout the coding region of the gene. Hum. Mutat. 2: 159-167, 1993. [PubMed: 8364584, related citations] [Full Text]

  348. Vulliamy, T. J., D'Urso, M., Battistuzzi, G., Estrada, M., Foulkes, N. S., Martini, G., Calabro, V., Poggi, V., Giordano, R., Town, M., Luzzatto, L., Persico, M. G. Diverse point mutations in the human glucose-6-phosphate dehydrogenase gene cause enzyme deficiency and mild or severe hemolytic anemia. Proc. Nat. Acad. Sci. 85: 5171-5175, 1988. [PubMed: 3393536, related citations] [Full Text]

  349. Vulliamy, T. J., Kaeda, J. S., Ait-Chafa, D., Mangerini, R., Roper, D., Barbot, J., Mehta, A. B., Athanassiou-Metaxa, M., Luzzatto, L., Mason, P. J. Clinical and haematological consequences of recurrent G6PD mutations and a single new mutation causing chronic nonspherocytic haemolytic anaemia. Brit. J. Haemat. 101: 670-675, 1998. [PubMed: 9674740, related citations] [Full Text]

  350. Vulliamy, T. J., Othman, A., Town, M., Nathwani, A., Falusi, A. G., Mason, P. J., Luzzatto, L. Polymorphic sites in the African population detected by sequence analysis of the glucose-6-phosphate dehydrogenase gene outline the evolution of the variants A and A-. Proc. Nat. Acad. Sci. 88: 8568-8571, 1991. [PubMed: 1924316, related citations] [Full Text]

  351. Vulliamy, T. J., Wanachiwanawin, W., Mason, P. J., Luzzatto, L. G6PD Mahidol, a common deficient variant in South East Asia is caused by a (163)glycine-to-serine mutation. Nucleic Acids Res. 17: 5868, 1989. [PubMed: 2503817, related citations] [Full Text]

  352. Vulliamy, T. J. Personal Communication. London, England 1989.

  353. Vulliamy, T., Mason, P., Luzzatto, L. The molecular basis of glucose-6-phosphate dehydrogenase deficiency. Trends Genet. 8: 138-143, 1992. [PubMed: 1631957, related citations] [Full Text]

  354. Vulliamy, T., Rovira, A., Yusoff, N., Colomer, D., Luzzatto, L., Vives-Corrons, J.-L. Independent origin of single and double mutations in the human glucose 6-phosphate dehydrogenase gene. Hum. Mutat. 8: 311-318, 1996. [PubMed: 8956035, related citations] [Full Text]

  355. Vuopio, P., Harkonen, M., Helske, T., Naeveri, H. Red cell glucose-6-phosphate dehydrogenase deficiency in Finland: characterization of a new variant with severe enzyme deficiency. Scand. J. Haemat. 15: 145-152, 1975. [PubMed: 1188317, related citations] [Full Text]

  356. Vuopio, P., Harkonen, M., Johnsson, P., Nuutinen, M. Red cell glucose-phosphate dehydrogenase deficiency in Finland. Ann. Clin. Res. 5: 168-178, 1973. [PubMed: 4749241, related citations]

  357. Waitz, R., Boivin, P., Oberling, F., Casenave, J. P., North, M. L., Mayer, S. Variante Gd(-) Strasbourg de la glucose-6-phosphate dehydrogenase. Nouv. Rev. Franc. Hemat. 10: 312-314, 1970. [PubMed: 4246285, related citations]

  358. Weimer, T. A., Salzano, F. M., Hutz, M. H. Erythrocyte isozymes and hemoglobin types in a southern Brazilian population. J. Hum. Evol. 10: 319-328, 1981.

  359. Weimer, T. A., Schuler, L., Beutler, E., Salzano, F. M. Gd(+) Laguna, a new rare glucose-6-phosphate dehydrogenase variant from Brazil. Hum. Genet. 65: 402-404, 1984. [PubMed: 6693129, related citations] [Full Text]

  360. Weinreich, J., Busch, D., Gottstein, U., Schaefer, J., Rohr, J. Ueber zwei neue Faelle von hereditaerer nichtsphaerocytaerer haemolytischer Anaemie bei glucose-6-phosphat-dehydrogenase-Defekt in einer Nord Deutschen Familie. Klin. Wschr. 46: 146-149, 1968. [PubMed: 5667020, related citations] [Full Text]

  361. Welch, S. G., McGregor, I. A., Williams, K. A new variant of human erythrocyte G6PD occurring at a high frequency amongst the population of two villages in The Gambia, West Africa. Hum. Genet. 40: 305-309, 1978. [PubMed: 631849, related citations] [Full Text]

  362. Westring, D. W., Pisciotta, A. V. Anemia, cataracts, and seizures in patients with glucose-6-phosphate dehydrogenase deficiency. Arch. Intern. Med. 118: 385-390, 1966. [PubMed: 5925237, related citations]

  363. WHO. Nomenclature of glucose-6-phosphate dehydrogenase in man. Bull. World Health Organ. 36: 319-322, 1967. Note: See Also: Canad. Med. Assoc. J. 97: 422-424, 1967. [PubMed: 5299754, related citations]

  364. WHO. Standardization of procedures for the study of glucose-6-phosphate dehydrogenase: report of a WHO Scientific Group. World Health Organ. Tech. Rep. Ser. 366: 1-53, 1967. [PubMed: 4963040, related citations]

  365. Wilson, W. W. Congenital hemolytic anemia due to a deficiency of glucose 6-phosphate dehydrogenase. Rocky Mt. Med. J. 73: 160-162, 1976.

  366. Witt, I., Yoshioka, S. Biochemical characterization of a glucose-6-phosphate dehydrogenase variant with favism: G-6-PD Zaehringen. Klin. Wschr. 50: 205-209, 1972. [PubMed: 5011553, related citations] [Full Text]

  367. Wong, P. W. K., Shih, L.-Y., Hsia, D. Y. Y. Characterization of glucose-6-phosphate dehydrogenase among Chinese. Nature 208: 1323-1324, 1965. [PubMed: 5870191, related citations] [Full Text]

  368. Yermakov, N., Tokarev, J., Chernjak, N., Schoenian, G., Grieger, M., Guckler, G., Jacobasch, G., Mahmudova, M., Bahramov, S. New stable mutant Gd(-) variants: G6PD Tashkent and G6PD Nucus: molecular basis of hereditary enzyme deficiency. Acta Biol. Med. Ger. 40: 559-562, 1981. [PubMed: 7315103, related citations]

  369. Yoshida, A., Baur, E. W., Motulsky, A. G. A Philippino glucose-6-phosphate dehydrogenase variant (G6PD Union) with enzyme deficiency and altered substrate specificity. Blood 35: 506-513, 1970. [PubMed: 4392654, related citations]

  370. Yoshida, A., Beutler, E., Motulsky, A. G. Table of human glucose-6-phosphate dehydrogenase variants. Bull. WHO 45: 243-253, 1971. [PubMed: 5316621, related citations]

  371. Yoshida, A., Beutler, E. Human glucose-6-phosphate dehydrogenase variants: a supplementary tabulation. Ann. Hum. Genet. 41: 347-355, 1978. [PubMed: 626479, related citations] [Full Text]

  372. Yoshida, A., Stamatoyannopoulos, G., Motulsky, A. G. Negro variant of glucose-6-phosphate dehydrogenase deficiency (A-) in man. Science 155: 97-99, 1967. [PubMed: 6015571, related citations] [Full Text]

  373. Yoshida, A., Takizawa, T., Prchal, J. T. RFLP of the X chromosome-linked glucose-6-phosphate dehydrogenase locus in blacks. Am. J. Hum. Genet. 42: 872-876, 1988. [PubMed: 2897162, related citations]

  374. Yoshida, A., Takizawa, T. The same extra FokI cleavage site exists in glucose-6-phosphate dehydrogenase variants A(+) and A(-). Am. J. Hum. Genet. 43: 131-133, 1988. [PubMed: 2840819, related citations]

  375. Yoshida, A. A single amino acid substitution (asparagine to aspartic acid) between normal (B plus) and the common Negro variant (A plus) of human glucose-6-phosphate dehydrogenase. Proc. Nat. Acad. Sci. 57: 835-840, 1967. [PubMed: 16591538, related citations] [Full Text]

  376. Yoshida, A. Human glucose-6-phosphate dehydrogenase: purification and characterization of Negro type variant (A+) and comparison with normal enzyme (B+). Biochem. Genet. 1: 81-99, 1967. [PubMed: 4388132, related citations] [Full Text]

  377. Yoshida, A. Amino acid substitution (histidine to tyrosine) in a glucose-6-phosphate dehydrogenase variant (G6PD Hektoen) associated with over-production. J. Molec. Biol. 52: 483-490, 1970. [PubMed: 5492291, related citations] [Full Text]

  378. Yoshida, A. Personal Communication. Duarte, Calif. 2/26/1996.

  379. Zuo, L., Chen, E., Du, C. S., Chang, C. N., Chiu, D. T. Y. Genetic study of Chinese G6PD variants by direct PCR sequencing. (Abstract) Blood 76 (suppl. 1): 51A, 1990.


Carol A. Bocchini - updated : 10/24/2013
Patricia A. Hartz - updated : 7/11/2013
Ada Hamosh - updated : 1/6/2010
Cassandra L. Kniffin - updated : 2/12/2009
Marla J. F. O'Neill - updated : 11/17/2008
Cassandra L. Kniffin - updated : 1/14/2008
Marla J. F. O'Neill - updated : 4/12/2007
Cassandra L. Kniffin - updated : 7/14/2006
Cassandra L. Kniffin - updated : 6/26/2006
Cassandra L. Kniffin - updated : 3/9/2006
Marla J. F. O'Neill - updated : 1/18/2006
Victor A. McKusick - updated : 12/16/2005
Victor A. McKusick - updated : 1/11/2005
Victor A. McKusick - updated : 1/3/2005
Victor A. McKusick - updated : 8/19/2003
Victor A. McKusick - updated : 4/24/2003
Victor A. McKusick - updated : 3/4/2003
Victor A. McKusick - updated : 1/14/2003
Victor A. McKusick - updated : 10/22/2002
Victor A. McKusick - updated : 9/12/2002
Victor A. McKusick - updated : 4/23/2002
Victor A. McKusick - updated : 4/4/2002
Deborah L. Stone - updated : 10/8/2001
Ada Hamosh - updated : 9/24/2001
Ada Hamosh - updated : 8/27/2001
Victor A. McKusick - updated : 7/13/2000
Victor A. McKusick - updated : 4/26/2000
Victor A. McKusick - updated : 3/15/1999
Victor A. McKusick - updated : 11/12/1998
Victor A. McKusick - updated : 10/13/1998
Victor A. McKusick - updated : 6/25/1998
Victor A. McKusick - updated : 3/24/1998
Michael J. Wright - updated : 9/25/1997
Victor A. McKusick - updated : 9/19/1997
Victor A. McKusick - updated : 3/18/1997
Moyra Smith - updated : 11/12/1996
Alan F. Scott - updated : 12/14/1995
Creation Date:
Victor A. McKusick : 7/6/1987
carol : 09/05/2023
carol : 08/30/2023
alopez : 08/09/2023
alopez : 04/28/2023
carol : 11/21/2022
carol : 04/01/2022
carol : 03/25/2022
carol : 03/04/2022
carol : 08/05/2019
carol : 06/20/2019
carol : 05/11/2018
carol : 05/24/2017
carol : 11/08/2016
carol : 09/12/2016
carol : 09/09/2016
carol : 07/28/2016
carol : 06/05/2015
alopez : 5/22/2015
alopez : 5/21/2015
carol : 9/4/2014
carol : 8/30/2014
carol : 6/18/2014
carol : 3/11/2014
joanna : 12/9/2013
mgross : 10/25/2013
carol : 10/25/2013
carol : 10/24/2013
carol : 10/24/2013
carol : 10/24/2013
mgross : 7/11/2013
carol : 4/18/2013
terry : 3/14/2013
terry : 12/20/2012
alopez : 3/5/2012
mgross : 7/7/2011
carol : 4/7/2011
terry : 11/8/2010
terry : 10/12/2010
alopez : 1/19/2010
terry : 1/6/2010
terry : 6/12/2009
terry : 3/27/2009
terry : 3/27/2009
wwang : 2/19/2009
ckniffin : 2/12/2009
wwang : 11/24/2008
terry : 11/21/2008
terry : 11/17/2008
wwang : 1/31/2008
ckniffin : 1/14/2008
wwang : 4/18/2007
terry : 4/12/2007
alopez : 2/1/2007
carol : 7/21/2006
carol : 7/21/2006
ckniffin : 7/14/2006
wwang : 7/13/2006
ckniffin : 6/26/2006
wwang : 5/25/2006
ckniffin : 3/9/2006
wwang : 1/18/2006
wwang : 12/28/2005
terry : 12/16/2005
terry : 2/7/2005
tkritzer : 1/21/2005
terry : 1/11/2005
wwang : 1/6/2005
terry : 1/3/2005
terry : 5/21/2004
joanna : 3/17/2004
carol : 2/23/2004
cwells : 11/7/2003
cwells : 11/7/2003
mgross : 8/21/2003
terry : 8/19/2003
carol : 4/24/2003
terry : 4/24/2003
cwells : 3/11/2003
terry : 3/4/2003
carol : 1/22/2003
tkritzer : 1/17/2003
terry : 1/14/2003
tkritzer : 10/29/2002
tkritzer : 10/24/2002
terry : 10/22/2002
carol : 9/18/2002
tkritzer : 9/13/2002
tkritzer : 9/13/2002
tkritzer : 9/12/2002
tkritzer : 9/12/2002
carol : 7/2/2002
alopez : 5/3/2002
terry : 4/23/2002
cwells : 4/15/2002
cwells : 4/9/2002
terry : 4/4/2002
carol : 10/8/2001
alopez : 9/25/2001
terry : 9/24/2001
alopez : 8/30/2001
terry : 8/27/2001
alopez : 7/21/2000
terry : 7/13/2000
mcapotos : 5/24/2000
mcapotos : 5/19/2000
terry : 4/26/2000
carol : 9/10/1999
joanna : 6/18/1999
carol : 5/27/1999
carol : 3/18/1999
terry : 3/15/1999
carol : 11/19/1998
terry : 11/12/1998
carol : 10/20/1998
carol : 10/18/1998
terry : 10/13/1998
dkim : 7/21/1998
alopez : 6/26/1998
terry : 6/25/1998
alopez : 3/31/1998
terry : 3/24/1998
dholmes : 2/10/1998
alopez : 1/14/1998
terry : 1/12/1998
terry : 1/12/1998
dholmes : 1/9/1998
alopez : 11/10/1997
alopez : 11/10/1997
alopez : 11/10/1997
terry : 9/19/1997
alopez : 7/28/1997
terry : 7/9/1997
mark : 7/7/1997
alopez : 6/27/1997
alopez : 6/27/1997
alopez : 6/27/1997
alopez : 6/27/1997
alopez : 6/27/1997
joanna : 6/23/1997
carol : 6/23/1997
mark : 6/16/1997
mark : 3/18/1997
terry : 3/11/1997
terry : 1/27/1997
jamie : 1/21/1997
terry : 1/14/1997
joanna : 1/1/1997
terry : 11/19/1996
mark : 11/12/1996
mark : 9/18/1996
mark : 7/9/1996
mark : 3/31/1996
terry : 3/21/1996
joanna : 3/15/1996
mark : 3/1/1996
terry : 3/1/1996
mark : 2/19/1996
terry : 2/15/1996
terry : 2/6/1996
mark : 1/17/1996
mark : 1/17/1996
joanna : 12/18/1995
mark : 12/15/1995
terry : 12/14/1995
mark : 7/19/1995
carol : 2/27/1995
davew : 8/5/1994
jason : 7/1/1994
pfoster : 4/22/1994
terry : 4/20/1994

* 305900

GLUCOSE-6-PHOSPHATE DEHYDROGENASE; G6PD


HGNC Approved Gene Symbol: G6PD

Cytogenetic location: Xq28     Genomic coordinates (GRCh38): X:154,531,390-154,547,569 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
Xq28 {Resistance to malaria due to G6PD deficiency} 611162 3
Hemolytic anemia, G6PD deficient (favism) 300908 X-linked 3

TEXT

Description

Glucose-6-phosphate dehydrogenase (G6PD; EC 1.1.1.49) plays a key role in the production of ribose 5-phosphate and the generation of NADPH in the hexose monophosphate pathway. Because this pathway is the only NADPH-generation process in mature red cells, which lack the citric acid cycle, a genetic deficiency of G6PD (300908) is often associated with adverse physiologic effects (summary by Takizawa et al., 1986).


Cloning and Expression

Takizawa et al. (1986) cloned G6PD from a human hepatoma cDNA library. The deduced 531-amino acid protein has a molecular mass of 58 kD. Cappellini and Fiorelli (2008) stated that the G6PD protein contains 515 amino acids.


Gene Structure

Martini et al. (1986) determined that the human G6PD gene has 13 exons and spans 18 kb. The protein-coding region is divided into 12 segments, ranging from 12 to 236 bp, and an intron is present in the 5-prime untranslated region. The major 5-prime end of mature G6PD mRNA in several cell lines is located 177 bp upstream of the translation-initiating codon. Comparison of the promoter region of G6PD and 10 other housekeeping enzyme genes confirmed the presence of common features. In particular, in 8 cases in which a 'TATA' box was present, a conserved sequence of 25 bp was seen immediately downstream.

Chen et al. (1991) determined the sequence of 20,114 bp of human DNA including the G6PD gene. The region included a prominent CpG island, starting about 680 nucleotides upstream of the transcription initiation site, extending about 1,050 nucleotides downstream of the initiation site, and ending at the start of the first intron. The transcribed region from the initiation site to the poly(A) addition site covered 15,860 bp. The sequence of the 13 exons agreed with the published cDNA sequence and, for the 11 exons tested, with the corresponding sequence in a yeast artificial chromosome (YAC). Sixteen Alu sequences constituted 24% of the total sequence tract. Four were outside the borders of the mRNA transcript of the gene; all of the others were found in a large (9,858 bp) intron between exons 2 and 3.

The Japanese pufferfish Fugu rubripes is a useful model for the comparative study of vertebrate genomes because of the compact nature of its genome. Since the Fugu genome is approximately 8 times smaller than that of mammals, most genes should be more compact. To test this hypothesis, Mason et al. (1995) cloned and sequenced the G6PD gene from Fugu and compared it to the corresponding human gene. The intron/exon structure of the 2 genes was identical throughout the protein coding regions. Intron 2 is also the largest intron in both species. However, they found that the Fugu gene was 4 times smaller than the human gene; the difference was accounted for by the fact that the pufferfish gene has smaller introns. Mason et al. (1995) constructed a molecular phylogeny for 10 G6PD protein sequences. The sequences fell in the expected arrangement based on established phylogenetic relationships, with the Plasmodium falciparum sequence diverging most widely.

Fusco et al. (2012) stated that the G6PD gene, which is transcribed in the telomeric direction, partly overlaps the IKBKG gene (300248), which is transcribed in the centromeric direction. The genes share a conserved promoter region that has bidirectional housekeeping activity. In addition, intron 2 of the G6PD gene contains an alternate promoter for the IKBKG gene only. Fusco et al. (2012) determined that the region containing the G6PD gene and the 5-prime end of the IKBKG gene contains Alu elements.


Evolution

Notaro et al. (2000) showed that an evolutionary analysis is a key to understanding the biology of a housekeeping gene such as G6PD. From the alignment of the amino acid sequence of 52 G6PD species from 42 different organisms, they found a striking correlation between the amino acid replacements that cause G6PD deficiency in humans and the sequence conservation of G6PD. Two-thirds of such replacements were found in highly and moderately conserved (50 to 99%) amino acids; relatively few were located in fully conserved amino acids (where they might be lethal) or in poorly conserved amino acids (where presumably they simply would not cause G6PD deficiency). The findings were considered consistent with the notion that all human mutants have residual enzyme activity and that null mutations are lethal at some stage of development. Comparing the distribution of mutations in the human housekeeping gene with evolutionary conservation is a useful tool for pinpointing amino acid residues important for the stability or the function of the corresponding protein.


Mapping

Childs et al. (1958) determined that the G6PD gene resides on the X chromosome.

From study of radiation-induced segregants (irradiated human cells 'rescued' by fusion with hamster cells), Goss and Harris (1977) showed that the order of 4 loci on the X chromosome is PGK: alpha-GAL: HPRT: G6PD and that the 3 intervals between these 4 loci are, in relative terms, 0.33, 0.30, and 0.23.

Studying X-autosome translocations in somatic cell hybrids, Pai et al. (1980) showed that a breakpoint at the junction of Xq27-q28 separates HPRT from G6PD. G6PD is distally situated at Xq28. They localized HPRT to the segment between Xq26 and Xq27.

That G6PD is X-linked in the mouse was supported by Epstein's finding (1969) that oocytes of XO females have half as much G6PD as do oocytes of XX female mice. The level of lactate dehydrogenase was the same. Epstein's conclusion was that the G6PD gene is X-linked in the mouse, that synthesis occurs in the oocyte and is dosage-dependent, and that X inactivation does not occur in oocytes.


Gene Function

Ninfali et al. (1995) studied muscle expression of G6PD in normal individuals and in persons with G6PD deficiency of 3 types. They were prompted to undertake these studies because of patients with symptoms such as myalgia, cramps, and muscle weakness under conditions of stress, particularly physical exertion. All 3 variants--Mediterranean (305900.0006), Seattle-like (305900.0010), and G6PD A- (305900.0002)--showed the enzyme defect in muscle. A statistically significant relationship was found in the activity of G6PD in erythrocytes and muscle of male subjects. The results suggested to the authors that, for a given variant, the extent of the enzyme defect in muscle can be determined from the G6PD activity of erythrocytes, using an equation that they derived.

In studies in bovine aortic and human coronary artery endothelial cells, Leopold et al. (2007) demonstrated that aldosterone decreased G6PD expression and activity, resulting in increased oxidant stress and decreased nitric oxide levels, similar to what is observed in G6PD-deficient endothelial cells. Aldosterone decreased G6PD expression by increasing expression of the cAMP-response element modulator (CREM; 123812), thereby inhibiting cAMP-response element binding protein (CREB; 123810)-mediated G6PD transcription. In vivo aldosterone infusion in mice decreased vascular G6PD expression and impaired vascular reactivity; these effects were abrogated by spironolactone or vascular gene transfer of G6pd. Leopold et al. (2007) concluded that aldosterone induces a G6PD-deficient phenotype to impair endothelial function.


Population Genetics

Different variants of G6PD are found in high frequency in African, Mediterranean, and Asiatic populations (Porter et al., 1964), and heterozygote advantage vis-a-vis malaria (Luzzatto et al., 1969) has been invoked to account for the high frequency of the particular alleles in particular populations.


Molecular Genetics

The variety of forms of the G6PD enzyme is great (Yoshida et al., 1971; Beutler and Yoshida, 1973; Yoshida and Beutler, 1978). The World Health Organization (WHO (1967, 1967)) gave its attention to problems of nomenclature and standard procedures for study. The demonstrated polymorphism at this X-linked locus rivals that of the autosomal loci for the polypeptide chains of hemoglobin. As in the latter instance, single amino acid substitution has been demonstrated as the basis of the change in the G6PD molecule resulting from mutation (Yoshida et al., 1967).

The G6PD variants have been divided into 5 classes according to the level of enzyme activity: class 1--enzyme deficiency with chronic nonspherocytic hemolytic anemia; class 2--severe enzyme deficiency (less than 10%); class 3--moderate to mild enzyme deficiency (10-60%); class 4--very mild or no enzyme deficiency (60%); class 5--increased enzyme activity. Mutations causing nonspherocytic hemolytic anemia are clustered near the carboxy end of the enzyme, in the region between amino acids 362 and 446, whereas most of the clinically mild mutations are located at the amino end of the molecule. As the intragenic defects have been identified, many variants that were thought to be unique have been found to be identical on sequence analysis. This finding should not be surprising inasmuch as the methods of biochemical characterization are not very accurate, particularly when dealing with mutant enzymes that are unstable. For example, although the patients were unrelated, G6PD Marion, G6PD Gastonia, and G6PD Minnesota had the same val213-to-leu substitution; and G6PD Nashville and G6PD Anaheim were found to have the same arg393-to-his substitution (Beutler et al., 1991).

The frequencies of low-activity alleles of G6PD in humans are highly correlated with the prevalence of malaria (see 611162). These deficiency alleles are thought to provide reduced risk for infection by the Plasmodium parasite and are maintained at high frequency despite the illnesses that they cause. Haplotype analysis of A- (305900.0002) and Mediterranean (Med) (305900.0006) mutations at this locus indicates that they had evolved independently and have increased in frequency at a rate that is too rapid to be explained by random genetic drift. Tishkoff et al. (2001) used statistical modeling to demonstrate that the A- allele arose within a past 3840 to 11,760 years and the Med allele arose within the past 1600 to 6640 years. Tishkoff et al. (2001) concluded that their results support the hypothesis that malaria has had a major impact on humans only since the introduction of agriculture within the past 10,000 years and provide a striking example of the signature of selection on the human genome.

That resistance to severe malaria is the basis of the high frequency of G6PD deficiency and that both hemizygotes and heterozygotes enjoy an advantage was established by Ruwende et al. (1995) in 2 large case-control studies of more than 2,000 African children. They found that the common African form of G6PD deficiency (G6PD A-; 305900.0002) was associated with a 46 to 58% reduction in risk of severe malaria for both female heterozygotes and male hemizygotes. A mathematical model incorporating the measured selective advantage against malaria suggested that a counterbalancing selective disadvantage, associated with this enzyme deficiency, has retarded its rise in frequency in malaria-endemic regions.

Sansone et al. (1981) described 6 G6PD variants in Italian males, all associated with enzyme deficiency and 2 with signs of hemolysis. They provided a useful map of 19 sporadic G6PD variants found in Italy. They mapped to regions where the common forms of G6PD deficiency are frequent.

Hitzeroth and Bender (1981) found an increasing frequency of apparent BB homozygotes with increasing age of groups of South African blacks studied. They suggested that this represents selection against A- cell lines in heterozygotes and speculated further that malaria is the underlying selective agent.

Mohrenweiser and Neel (1981) identified thermolabile variants of lactate dehydrogenase B, glucosephosphate isomerase, and glucose-6-phosphate dehydrogenase. None was detectable as a variant by standard electrophoretic techniques. All were inherited. Beutler (1983) hypothesized that the marked differences in the extent to which various tissues manifest the deficiency state in various enzymopathies including G6PD deficiency may be related to tissue-to-tissue differences in proteases. Mutation may produce changes in susceptibility of the enzyme to proteases.

Data on gene frequencies of allelic variants were tabulated by Roychoudhury and Nei (1988).

Vulliamy et al. (1988) cloned and sequenced 7 mutant G6PD alleles. A single point mutation in the African variant G6PD A (305900.0001) does not result in deficiency of the enzyme. The other 6 mutants were all associated with enzyme deficiency. Single point mutations were identified in G6PD Mediterranean (305900.0006), G6PD Metaponto (305900.0007), G6PD Ilesha (305900.0004), G6PD Chatham (305900.0003), G6PD Santiago de Cuba (305900.0009), and G6PD Matera (an example of A-; 305900.0002).

By use of 14 unique sequence probes and 18 restriction enzymes, D'Urso et al. (1988) found a polymorphic silent mutation in the G6PD gene. A PstI site that maps to exon 10 was monomorphic in all British and Italian subjects studied, but was polymorphic in West African people. Specifically, it was absent from 22% of Nigerian X chromosomes. By sequence analysis, D'Urso et al. (1988) showed that the absence of this PstI site resulted from a G-to-A replacement at position 1116, corresponding to the third base of a glutamine codon (see 305900.0017). No amino acid change was produced in the protein. Yoshida et al. (1988) reported 2 RFLPs of the G6PD locus with high frequency in blacks and showed statistically significant linkage disequilibrium between the A+/B+ types and 1 of the RFLPs at the G6PD locus.

Vulliamy et al. (1988) found a striking predominance of C-to-T transitions among the G6PD mutations, with GC doublets involved in 4 of the 7 cases. It has been found that even in the same population, more than 1 G6PD variant is present. For example, in the island of Sardinia, extensive clinical and biochemical studies identified 4 different G6PD variants. De Vita et al. (1989) cloned and sequenced the 4 G6PD variants and found that at the molecular level there were only 2 mutations. The first mutation had an asp282-to-his change resulting from a GAT-to-CAT change in exon 8. This mutation caused the G6PD Seattle-like phenotype, a relatively mild form of G6PD deficiency (see 305900.0010). The other 3 variants were accompanied by very severe G6PD deficiency. All 3 had a ser188-to-phe change resulting from a TCC-to-TTC transition. This is the same change as that in G6PD Mediterranean (305900.0006). These 3 Sardinian variants also had a silent mutation in exon 11 with a change of TAC-to-TAT, both of which encode tyrosine at amino acid 437. These findings indicate that some G6PD-deficient variants identified only on the basis of their biochemical characteristics may not correspond to different mutations in the G6PD gene. The variations may be due to posttranscriptional or posttranslational modifications of the enzyme; whether the modifications are due to mutations in a tightly linked gene or to noninherited physiologic changes could not be distinguished with the data available. Study of families in which different forms of G6PD Mediterranean segregate suggested that the biochemical characteristics are transmitted in the family along with the enzyme deficiency, thus favoring the first hypothesis.

In a study of an unselected sample of 1,524 schoolboys from the province of Matera (Lucania) in southern Italy, Calabro et al. (1990) found that although the most frequent form of G6PD deficiency was G6PD Mediterranean, an extraordinary number of other forms existed. The overall rate of G6PD deficiency was 2.6%. The frequency ranged from 7.2% on the Ionian coast to zero on the eastern side of the Lucanian Apennines.

Kay et al. (1992) analyzed the evolution of the G6PD gene by examining the DNA samples from 54 male African Americans for G6PD A+ (305900.0001), G6PD A- (305900.0002), and G6PD B and for polymorphisms in intron 5 (PvuII), at nucleotide 1311 (305900.0018), and at nucleotide 1116 (305900.0017). They concluded from these and their previous studies that G6PD B is the most ancient genotype. The nucleotide 1311 mutation, with its worldwide distribution, probably occurred next. The PstI mutation, limited to Africans, probably arose next and is more ancient than the A+ mutation, which occurred in a gene without either the PstI or the 1311 mutation. G6PD A- (202A/376G) is the most recent mutation and is still in linkage disequilibrium with all of the sites. It presumably occurred in an individual with both the A+ and PvuII mutations.

Chiu et al. (1993) reported molecular characterization of the defects in 43 G6PD-deficient Chinese males whose G6PD had been well characterized biochemically. Among the 43 samples, they identified 5 different nucleotide substitutions: 1388G-A (arg to his; 305900.0029); 1376G-T (arg to leu; 305900.0021); 1024C-T (leu to phe; 305900.0046); 392G-T (gly to val; 305900.0045); and 95A-G (his to arg; 305900.0044). The 5 substitutions accounted for 36 of the 43 samples; none of these substitutions had been reported in non-Asians. The substitutions at nucleotides 392 and 1024 were new findings. The substitutions at nucleotides 1376 and 1388 accounted for over one-half of the samples.

Vulliamy et al. (1993) tabulated 58 different mutations in the G6PD gene that account for 97 named G6PD variants. The mutations were almost exclusively missense mutations, causing single amino acid substitutions. They were spread throughout the coding region of the gene, although there appeared to be a clustering of mutations that caused a more severe clinical phenotype towards the 3-prime end of the gene. The absence of large deletions, frameshift mutations, and nonsense mutations was considered consistent with the notion that a total lack of G6PD activity would be lethal.

Miwa and Fujii (1996) listed the mutations responsible for about 78 G6PD variants.

Mason (1996) reviewed information on the G6PD enzyme and on mutations in the gene. A map of 515 amino acids showing the location of mutations, including double mutations, was provided.

Filosa et al. (1996) analyzed fractionated blood cells in 4 heterozygotes for the class 1 G6PD mutations G6PD Portici (305900.0008) and G6PD Bari (1187G-T). In erythroid, myeloid, and lymphoid cell lineages there was a significant excess of G6PD-normal cells, suggesting that a selective mechanism operates at the level of pluripotent blood stem cells. They concluded that their studies provided evidence that severe G6PD deficiency adversely affects the proliferation or survival of nucleated blood cells.

Liu et al. (1997) reported a method of determination of clonality using allele-specific PCR (ASPCR) to detect exonic polymorphisms in p55 (305360) and G6PD. They demonstrated a significant sex difference in allele frequencies in African Americans but not in Caucasians, and linkage disequilibrium for the p55 and G6PD alleles in Caucasians but not in African Americans.

Vulliamy et al. (1998) determined the causative mutation in 12 cases of G6PD deficiency associated with chronic nonspherocytic hemolytic anemia. In 11 cases, the mutation they found had previously been reported in unrelated individuals. These mutations comprised 7 different missense mutations and a 24-bp deletion, G6PD Nara (305900.0052), previously found in a Japanese boy. Repeated findings of the same mutations suggest that a limited number of amino acid changes can produce the chronic nonspherocytic hemolytic anemia phenotype and be compatible with normal development. They found 1 new mutation, G6PD Serres (305900.0051).

Cappadoro et al. (1998) presented evidence suggesting that early phagocytosis of G6PD-deficient erythrocytes parasitized by Plasmodium falciparum may explain malaria protection in G6PD deficiency.

Kwok et al. (2002) described a Web-accessible database of G6PD mutations. The relational database integrates up-to-date mutational and structural data from various databanks with biochemically characterized variants and their associated phenotypes obtained from published literature and a Favism website.

Barisic et al. (2005) identified 5 different mutations in the G6PD gene in 24 unrelated males with G6PD deficiency from the Dalmatian region of southern Croatia. The variants included Cosenza (305900.0059) (37.5% of patients), Mediterranean (305900.0006) (16.6%), Seattle (12.5%), Union (12.5%), Cassano (4.2%), and a novel variant, termed G6PD Split (305900.0059) (4.2%). The variants in 3 patients (12.5%) were uncharacterized.

Ninokata et al. (2006) identified G6PD deficiency in 9.8% of males and 10.4% of females among 345 healthy adults on Phuket island in southern Thailand. Although none of the individuals had molecular evidence of malaria infection, the findings suggested that malaria endemics had occurred in the past and that G6PD deficiency has been maintained as an advantageous genetic trait in this population. At least 5 different G6PD variants were identified, suggesting that several Asian ethnic groups, such as Burmese, Laotian, Cambodian, Thai, and Chinese, participated in establishing the current ethnic identity of the population of Phuket.

Jiang et al. (2006) identified 14 different mutations in the G6PD gene among 1,004 G6PD-deficient Chinese individuals comprising 11 ethnic groups. The variants varied in frequency across the ethnic groups and correlated geographically with historical patterns of malaria. The variants were different from those reported in African, European, and Indian populations. The most common variants in the Chinese population were G6PD Kaiping (R463H; 305900.0029) and G6PD Canton (R459L; 305900.0021), accounting for over 60% of G6PD-deficient individuals, and Gaohe (H32R; 305900.0044). In vitro functional expression studies in E. coli showed significantly decreased enzyme activity for all 3 mutant proteins. All 3 variants showed decreased Km for G6P, but whereas the Canton and Kaiping variants had increased Km for NADP+, the Gaohe variant showed decreased Km for NADP+, likely reflecting compensation in the latter variant. Jiang et al. (2006) concluded that residues arg459 and arg463 play an important role in anchoring NADP+ to the catalytic domain of the enzyme.


Genotype/Phenotype Correlations

Miwa and Fujii (1996) stated that most of the class 1 G6PD variants associated with chronic hemolysis have the mutations surrounding either the substrate- or NADP-binding site.

Costa et al. (2000) pointed out that G6PD mutants causing class 1 variants (the most severe forms of the disease) cluster within exon 10, in a region that, at the protein level, is believed to be involved in dimerization. They identified a class 1 variant mapping to exon 8 (305900.0053).


Animal Model

Longo et al. (2002) crossed mouse chimeras from embryonic stem cells in which the G6pd gene had been targeted with normal females. First-generation G6pd heterozygotes born from this cross were essentially normal; their tissues demonstrated strong selection for cells with the targeted G6pd allele on the inactive X chromosome. When these first-generation heterozygous females were bred to normal males, only normal G6pd mice were born. There were 3 reasons for this: hemizygous G6pd male embryos' development was arrested from embryonic day 7.5, the time of onset of blood circulation, and they died by embryonic day 10.5; heterozygous G6pd females showed abnormalities from embryonic day 8.5, and died by embryonic day 11.5; and severe pathologic changes were present in the placenta of both G6pd hemizygous and heterozygous embryos. Thus, G6PD is not indispensable for early embryonic development; however, severe G6PD deficiency in the extraembryonic tissues (consequent on selective inactivation of the normal paternal G6PD allele) impairs the development of the placenta and causes death of the embryo. Most importantly, G6PD is indispensable for survival when the embryo is exposed to oxygen through its blood supply.

In ischemia-reperfusion experiments on isolated mouse hearts, Jain et al. (2004) demonstrated that G6pd is rapidly activated without a change in G6pd protein levels. G6pd -/- hearts had greatly impaired cardiac relaxation and contractile performance, associated with depletion of total glutathione stores and impaired generation of reduced glutathione, compared to wildtype hearts. Increased ischemia-reperfusion injury was reversed by antioxidant treatment but unaffected by supplementation of ribose stores. Jain et al. (2004) concluded that G6PD is an essential myocardial antioxidant enzyme, required for maintaining cellular glutathione levels and protecting against oxidative stress-induced cardiac dysfunction during ischemia-reperfusion.


History

Polymorphism at the G6PD locus has made it a useful X-chromosome marker, like the colorblindness and Xg blood group loci; close linkage of the colorblindness loci, the G6PD locus, and the hemophilia A locus (Adam et al., 1967; Boyer and Graham, 1965) has been demonstrated. Also, as a biochemical phenotype identifiable at the cellular level, G6PD variants have been useful in somatic cell genetics, permitting, for example, one of the critical proofs in man of the Lyon hypothesis (Davidson et al., 1963).

The relative stability of the X chromosome during evolution has been shown by the fact that the G6PD locus is X-borne also in a number of other species (Ohno, 1967). G6PD and HPRT are linked in the Chinese hamster (Rosenstraus and Chasin, 1975) and presumably are on the X chromosome as in man. By study of cell hybrids, Shows et al. (1976) found that HPRT and G6PD are closely linked in the Muntjac deer. Smith et al. (1976) found G6PD deficiency in a male Weimaraner dog, but were not able to do genetic studies. Alpha-GAL, HPRT, PGK and G6PD are X-linked in the rabbit, according to mouse-rabbit hybrid cell studies (Cianfriglia et al., 1979; Echard and Gillois, 1979). By comparable methods, Hors-Cayla et al. (1979) found them to be X-linked also in cattle. According to cell hybridization studies, HPRT, G6PD, and PGK are X-linked in the pig (Gellin et al., 1979) and in sheep (Saidi et al., 1979). Using pulsed field gel electrophoresis, Faust et al. (1992) demonstrated that, in the mouse, Gdx (312070), P3 (312090), and G6pd are physically linked to the X-linked visual pigment locus (Rsvp) within a maximal distance of 340 kb, while G6pd and f8 (300841) are approximately 900 kb apart.

Takizawa and Yoshida (1987) reported that the G6PD A+ gene has an A-to-G transition, resulting in the substitution of aspartic acid for asparagine as the 142nd amino acid from the N-terminus of the enzyme.

G6PD Hektoen is characterized by increased red cell enzyme activity. It is, therefore, a class 5 G6PD variant. It was first described by Dern et al. (1969). Yoshida (1970) thought that the variant peptide had replacement of histidine by tyrosine. Later, Yoshida (1996) was uncertain about this conclusion and stated that the basic defect remained to be identified.


ALLELIC VARIANTS 63 Selected Examples):

.0001   G6PD A+

G6PD, ASN126ASP
SNP: rs1050829, gnomAD: rs1050829, ClinVar: RCV000011073, RCV000011075, RCV000011109, RCV000079405, RCV000178823, RCV000307631, RCV000477820, RCV000761430, RCV000999876, RCV001095678, RCV001267359, RCV001375611, RCV002305425, RCV002305432, RCV002305707, RCV002305727, RCV002305732, RCV002305803, RCV002305807, RCV002305808, RCV002305836, RCV002305857

See Kirkman et al. (1964) and Yoshida et al. (1967). Vulliamy et al. (1988) found that the G6PD A variant is the same as 1 of the 2 variants identified in G6PD A- (305900.0002), i.e., asn126-to-asp. They noted that G6PD A, which is widely distributed in Africa, is not associated with deficiency of the enzyme.

Hirono and Beutler (1988) showed that a mutation responsible for the G6PD A- phenotype present in enzyme-deficient (300908) West African and American blacks occurred in a gene that produces the G6PD A+ phenotype. A substitution of guanine for adenine at nucleotide 376 (in exon 5) was found in all G6PD A+ and G6PD A- samples but in none of the G6PD B+ samples examined. Substitution of adenine for guanine at nucleotide 202 was found in 4 of 5 G6PD A- samples; this change is apparently responsible for the in vivo instability of the enzyme protein. Thus, the difference distinguishing the A and B forms of G6PD is the amino acid at residue 126 (see 305900.0002). Presumably as the result of alternative splicing, there is considerable heterogeneity among different G6PD cDNAs.

Both the variant A (with enzyme activity in the normal range, also called A) and the variant A- (associated with enzyme deficiency) have a frequency of about 0.2 in several African populations. Two restriction fragment length polymorphisms have also been found in people of African descent but not in other populations, whereas a silent mutation has been shown to be polymorphic in Mediterranean, Middle Eastern, African, and Indian populations. Vulliamy et al. (1991) reported 2 additional polymorphisms detected by sequence analysis, one in intron 7 and one in intron 8. Analysis of 54 African males for the 7 polymorphic sites clustered within 3 kb of the G6PD gene showed only 7 of the 128 possible haplotypes, thus indicating marked linkage disequilibrium. These data enabled Vulliamy et al. (1991) to suggest an evolutionary pathway for the different mutations, with only a single ambiguity. The mutation underlying the A variant is the most ancient and the mutation underlying the A- variant is the most recent. Since it seemed reasonable that the A- allele is subject to positive selection by malaria, whereas the other alleles are neutral, Vulliamy et al. (1991) suggested that G6PD may lend itself to the analysis of the role of random genetic drift and selection in determining allele frequencies within a single genetic locus in human populations.


.0002   G6PD A-

G6PD MATERA
G6PD BETICA
G6PD CASTILLA
G6PD DISTRITO FEDERAL
G6PD TEPIC
G6PD, VAL68MET, ASN126ASP
SNP: rs1050828, gnomAD: rs1050828, ClinVar: RCV000011073, RCV000011075, RCV000011109, RCV000011157, RCV000079404, RCV000079405, RCV000178823, RCV000224469, RCV000307631, RCV000477820, RCV000606259, RCV000761430, RCV000999820, RCV000999876, RCV001095678, RCV001095679, RCV001267358, RCV001267359, RCV001375611, RCV002305425, RCV002305432, RCV002305706, RCV002305707, RCV002305727, RCV002305732, RCV002305803, RCV002305806, RCV002305807, RCV002305808, RCV002305821, RCV002305836, RCV002305857, RCV003460495

Babalola et al. (1976) predicted that the A- mutation may have occurred in an individual carrying the A+ mutation. A black individual with the G6PD A- phenotype but no mutation at nucleotide 202 suggested that this individual may have another mutation that caused instability and thus deficiency of the enzyme. Yoshida and Takizawa (1988) presented evidence that the A- gene evolved by stepwise mutations through the A+ gene.

Vulliamy et al. (1988) cloned and sequenced 7 mutant G6PD alleles. A single point mutation in the African variant G6PD A does not result in deficiency of the enzyme. The other 6 mutants, including G6PD A-, were all associated with enzyme deficiency. Two different point mutations were found in G6PD A-, 1 of which was the same as that in G6PD A. See Yoshida et al. (1967). Hirono and Beutler (1988) demonstrated a substitution of methionine for valine at position 68 resulting from a G-to-A change at nucleotide 202 (in exon 4). The in vivo instability of the enzyme is the result of this change. The gene also has the change at amino acid 126 characteristic of G6PD A. See Vulliamy et al. (1988).

Beutler et al. (1989) performed haplotyping with 4 polymorphic restriction sites in the G6PD locus in DNA samples from 29 males with the G6PD A- phenotype and 14 males with a G6PD B phenotype. All G6PD A- subjects with the G6PD A- (202A/376G) genotype, regardless of population origin, shared identical haplotypes. The 5 populations screened were black (16), Puerto Rican (2), Mexican (2), white US (1), and Spanish (3). One G6PD A- male was of the 376G/680T genotype and 2 were of the 376G/968C genotype. One of the restriction sites is uncommon in the populations studied; thus, Beutler and Kuhl (1990) considered it likely that the G6PD A- mutation at nucleotide 202 arose relatively recently and in a single person.

Calabro et al. (1990) found this mutation, regarded as characteristically African, in an unselected sample of 1,524 schoolboys of the province of Matera in Southern Italy.

Beutler et al. (1991) found that 3 previously reported electrophoretically fast Mexican G6PD variants--G6PD Distrito Federal (Lisker et al., 1981), G6PD Tepic (Lisker et al., 1985), and G6PD Castilla (Lisker et al., 1977)--all showed the changes characteristic of G6PD A- (202A/376G) and had the haplotype characteristic of G6PD A- in Africa. G6PD Betica (Vives-Corrons and Pujades, 1982; Vives-Corrons et al., 1980), which is frequent in Spain, also had the same characteristics. Since the PvuII+ genotype is rare in Europe, the G6PD Betica mutation was presumably imported from Africa.

Hirono and Beutler (1988) found 2 other mutations that produced the G6PD A- phenotype: arg227-to-leu and leu323-to-pro. In both cases the mutations existed on the G6PD A background, i.e., the asn126-to-asp substitution.

Town et al. (1992) demonstrated that both the val68-to-met and the asn126-to-asp mutations found in G6PD A- are necessary to produce the G6PD-deficient phenotype (rather than the val68-to-met mutation having happened to arise in an A+ gene in the first instance). They approached the question by introducing G6PD B, A, A-, and G6PD val68-to-met in a bacterial expression system and analyzing their biochemical properties. With each of the 2 mutations alone, they found a slight decrease in both the specific activity and the yield of enzyme protein when compared to G6PD B. When both mutations were introduced together, there was a roughly additive effect on specific activity, but a much more drastic effect on enzyme yield which was reduced to 4% of normal. They inferred that the coexistence of the 2 mutations acted synergistically in causing instability of the enzyme. This would explain why a B- phenotype has only very rarely been observed. (Comparable results were produced when the replacement gln119-to-glu was combined with val68 to met.)

G6PD A- is the most common polymorphic variant associated with deficiency of G6PD in African populations, accounting for 20 to 40% of the affected population in western and central Africa; the most common nondeficient polymorphic variant in Africa is G6PD A. The G6PD A- mutation at position 68 alone has not been detected in any variant; this, together with further haplotyping analyses, led Vulliamy et al. (1992) to suggest that the nondeficient single mutant G6PD A is more ancient than the deficient double mutant G6PD A-.

Gomez-Gallego et al. (2000) performed structural studies on the doubly mutant G6PD A-. The changes they observed did not affect the active site of the mutant protein, since its spatial position was unmodified. The result of the structural changes was a loss of folding determinants, leading to a protein with decreased intracellular stability. Gomez-Gallego et al. (2000) suggested that the resultant protein was the cause of the enzyme deficiency in the red blood cell, which is unable to perform de novo protein synthesis.


.0003   G6PD CHATHAM

G6PD, ALA335THR
SNP: rs5030869, gnomAD: rs5030869, ClinVar: RCV000011081, RCV000180546, RCV000622381, RCV000757321, RCV000763202, RCV001000058, RCV003460451

Substitution of adenine for guanine at nucleotide 1003 leads to substitution of alanine by threonine at amino acid position 335 (Vulliamy et al., 1988). This mutation has been found in 2 unrelated Asian Indians and in a man from Syria and may be polymorphic. It causes class 2 enzyme derangement. No change in restriction sites has been found.

Mesbah-Namin et al. (2002) reported the first investigation of G6PD deficiency (300908) among the Mazandaranians of northern Iran. They analyzed the G6PD gene in 74 unrelated G6PD-deficient males with a history of favism. Molecular analysis revealed 3 different major polymorphic variants: G6PD Mediterranean (305900.0006) was found in 49 (66.2%), G6PD Chatham in 20 (27%), and G6PD Cosenza in 5 (6.75%) of the patients. The prevalence of G6PD Chatham in this Iranian population was the highest in the world. The distribution of the G6PD variants was more similar to that found in an Italian population than in other Middle Eastern countries.


.0004   G6PD ILESHA

G6PD, GLU156LYS
SNP: rs137852313, gnomAD: rs137852313, ClinVar: RCV000011082, RCV000991017, RCV002227031

See Usanga et al. (1977) and Luzzatto et al. (1979). Substitution of adenine for guanine at base 466 (in exon 5) leads to replacement of glutamic acid by lysine (Vulliamy et al., 1988). This sporadic class 3 mutation is associated with loss of a HinfI site.


.0005   G6PD MAHIDOL

G6PD, GLY163SER
SNP: rs137852314, gnomAD: rs137852314, ClinVar: RCV000011085, RCV000282708, RCV000507435, RCV000657881, RCV000763205, RCV002298440, RCV003466846

See Panich et al. (1972). A G-to-A change at base 487 (exon 6) leads to substitution of serine for glycine at amino acid 163 (Vulliamy, 1989). This mutation is polymorphic in Southeast Asia, causes class 2 enzyme derangement, and is associated with a new AluI site (Vulliamy et al., 1989). The same mutation was identified by Tang et al. (1992) in a Taiwanese in Taiwan.

Matsuoka et al. (2004) found that 11% of blood samples from persons in remote areas of Myanmar (former Burma) indicated G6PD deficiency. Taken together with data from a previous report (Iwai et al., 2001), these findings indicated that 91.3% of G6PD variants were G6PD Mahidol. The findings suggested that the Myanmar population is derived from homogeneous ancestries different from those of Thai, Malaysian, and Indonesian populations.

Louicharoen et al. (2009) investigated the effect of the G6PD-Mahidol 487A variant on human survival related to P. vivax and P. falciparum malaria in Southeast Asia. They showed that strong and recent positive selection has targeted the Mahidol variant over the past 1,500 years. The authors found that the G6PD-Mahidol variant reduces vivax, but not falciparum, parasite density in humans, which indicates that P. vivax has been a driving force behind the strong selective advantage conferred by this mutation.


.0006   G6PD MEDITERRANEAN

G6PD SASSARI
G6PD CAGLIARI
G6PD, SER188PHE
SNP: rs5030868, gnomAD: rs5030868, ClinVar: RCV000011086, RCV000011087, RCV000011088, RCV000079409, RCV000179363, RCV000445579, RCV000477810, RCV000623137, RCV000761429, RCV001250219, RCV001265539, RCV001528124, RCV002227060, RCV002305806, RCV002305807, RCV002305808, RCV002305849, RCV003925095

See Kirkman et al. (1964), Ben-Bassat and Ben-Ishay (1969), Lenzerini et al. (1969), Testa et al. (1980), and Morelli et al. (1984). A change from cytosine to thymine at base position 563 (in exon 6) causes a change from serine to phenylalanine in amino acid position 188 (Vulliamy et al., 1988). De Vita et al. (1989) found that G6PD Mediterranean, G6PD Sassari, and G6PD Cagliari have the same mutational change, resulting from a TCC-to-TTC mutation in exon 6. There is a second silent mutation of TAC-to-TAT at codon 437 in exon 11 (C-to-T at nucleotide 1311; see 305900.0018); both codons code for tyrosine. This mutation is a polymorphism, causes class 2 abnormality, and creates a new MboII site.

Beutler and Kuhl (1990) studied the distribution of the nucleotide polymorphism C1311T in diverse populations. Only 1 of 22 male subjects from Mediterranean countries who had the G6PD Mediterranean-563T genotype had a C at nucleotide 1311, which is the more frequent finding in this group. In contrast, both G6PD Mediterranean-563T males from the Indian subcontinent had the usual C at nucleotide 1311. Beutler and Kuhl (1990) interpreted these findings as suggesting that the same mutation at nucleotide 563 arose independently in Europe and in Asia.

Similar studies were done by Kurdi-Haidar et al. (1990) in 21 unrelated individuals with G6PD Mediterranean from Saudi Arabia, Iraq, Iran, Jordan, Lebanon, and Israel. All but 1 had the 563 mutation, and, of these, all but 1 had the C-to-T change at nucleotide 1311. Among another 24 unrelated Middle Eastern persons with normal G6PD activity, 4 had the silent mutation at position 1311 in the absence of the deficiency mutation at position 563. Kurdi-Haidar et al. (1990) concluded that most Middle Eastern subjects with the G6PD Mediterranean phenotype have the same mutation as that found in Italy; that the silent mutation is an independent polymorphism in the Middle East, with a frequency of about 0.13; and that the mutation leading to G6PD Mediterranean deficiency probably arose on a chromosome that already carried the silent mutation.

In Nepal, Matsuoka et al. (2003) tested 300 males for G6PD deficiency and identified 2 (0.67%) who were G6PD deficient. Compared with normal controls, G6PD activity was 12% and 26%, respectively. Both subjects had the 563C-T substitution of G6PD Mediterranean (ser188 to phe), and both had the silent 1311C-T change. A similar second change has been described in persons living in Mediterranean countries and Middle East countries. However, the form of G6PD Mediterranean found in India and Pakistan has no replacement at nucleotide 1311. Thus, the 2 subjects in Kathmandu, Nepal, would be closer to people in Middle East countries than people in India.

Corcoran et al. (1992) described a G6PD mutant biochemically indistinguishable from the common variety due to a C-to-T mutation at nucleotide 563. Instead, a C-to-T transition was found at nucleotide 592 in exon 6, changing an arginine residue to a cysteine residue only 10 amino acids downstream from the Mediterranean mutation. The new variant was named G6PD Coimbra (305900.0031).

Kaplan et al. (1997) presented data suggesting that the coexistence of Mediterranean type G6PD deficiency with the TA insertion polymorphism of the promoter of the UGT1A1 gene (191740.0011), which is associated with Gilbert syndrome (143500) in adults, is responsible for the development of neonatal hyperbilirubinemia. This is the most devastating clinical consequence of G6PD deficiency; it can be severe and result in kernicterus or even death. Kaplan et al. (1997) found that neither G6PD deficiency nor the polymorphism of UDP glucuronosyltransferase alone increased the incidence of neonatal hyperbilirubinemia, but in combination they did. The authors suggested that this gene interaction may serve as a paradigm of the interaction of benign genetic polymorphisms in the causation of disease.

Kaplan et al. (2001) reported 2 premature female neonates heterozygous for the G6PD Mediterranean mutation who presented with severe hyperbilirubinemia requiring exchange transfusions. Both had had normal G6PD biochemical screening tests.


.0007   G6PD METAPONTO

G6PD, ASP58ASN
SNP: rs137852315, ClinVar: RCV000011089, RCV002305426

Substitution of adenine for guanine at base 172 (exon IV) leads to a substitution of asparagine for aspartic acid at amino acid 58 (Vulliamy et al., 1988). The mutation was found in a sporadic, class 3 case, and no restriction site change was identified. See Calabro et al. (1990).


.0008   G6PD PORTICI

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, ARG393HIS
SNP: rs137852316, ClinVar: RCV000011090, RCV000030890, RCV000030891, RCV000066231, RCV000756196, RCV003480028

G6PD Portici has a G-to-A change at nucleotide 1178 of the G6PD gene, resulting in substitution of histidine for arginine at residue 393 (Filosa, 1989). The mutation was found in a sporadic case of class 1 deficiency (300908) and is not associated with an identified restriction site. In the full report, Filosa et al. (1992) described the kinetic characteristics of this G6PD variant (Portici) which was associated with chronic nonspherocytic hemolytic anemia.


.0009   G6PD SANTIAGO DE CUBA

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, GLY447ARG
SNP: rs137852317, ClinVar: RCV000011091, RCV000066233, RCV000790772, RCV002496322

Substitution of adenine for guanine as base number 1339 (exon 11) leads to substitution of arginine for glycine at amino acid position 447 (Vulliamy et al., 1988). This variant is associated with severe chronic hemolytic anemia (class 1; 300908). It was found in a sporadic case. A new PstI site was created, and this was used to show that it was a new mutation.


.0010   G6PD SEATTLE-LIKE

G6PD MODENA
G6PD, ASP282HIS
SNP: rs137852318, gnomAD: rs137852318, ClinVar: RCV000011092, RCV000011093, RCV000180200, RCV000757322, RCV000763204, RCV001166655, RCV003466847, RCV003904827

See Lenzerini et al. (1969) and Rattazzi et al. (1969). De Vita et al. (1989) found that G6PD Seattle-like, which produces a relatively mild phenotype, has substitution of histidine for aspartic acid at amino acid 282, resulting from a GAT-to-CAT change in exon 8. Cappellini et al. (1994) found the same variant in an Italian man from the Po delta and designated it G6PD Modena before finding that it had the same mutation as that in G6PD Seattle-like. They stated that the G-to-C transition was at nucleotide 844 in exon 8.


.0011   G6PD HARILAOU

G6PD, PHE216LEU
SNP: rs137852319, gnomAD: rs137852319, ClinVar: RCV000011094, RCV002305427

Town et al. (1990) described G6PD Harilaou in a Greek boy with severe hemolytic anemia. Poggi (1989) found a T-to-G change at nucleotide 648 that leads to substitution of leucine for phenylalanine at residue 216.


.0012   G6PD IOWA

G6PD IOWA CITY
G6PD SPRINGFIELD
G6PD WALTER REED
G6PD, LYS386GLU
SNP: rs137852320, ClinVar: RCV000011096, RCV000011097, RCV000011098, RCV000011099, RCV000294855, RCV002305428

See Beutler et al. (1986). Hirono et al. (1989) demonstrated an A-to-G substitution at nucleotide 1156, resulting in substitution of glutamic acid for lysine at amino acid 386. This variant G6PD, as well as G6PD Beverly Hills, Tomah, Riverside, and some others, is unstable in the presence of 10 microM NADP+ (where normal G6PD is stable) but is reactivated by 200 microM NADP+. G6PD Tomah, Iowa and Beverly Hills have amino acid substitutions at positions 385, 386, and 387, respectively; G6PD Riverside, with a substitution at position 410, shows weak reactivation by NADP+. These observations, together with the fact that these amino acids are highly conserved, led Hirono et al. (1989) to propose that they are in the region of the molecule involved in NADP+ binding.


.0013   G6PD BEVERLY HILLS

G6PD, ARG387HIS
SNP: rs137852321, ClinVar: RCV000011100, RCV001857328, RCV003466848

Hirono et al. (1989) demonstrated a G-to-A mutation at nucleotide 1160, causing substitution of histidine for arginine-387. The mutation destroyed an HhaI site.


.0014   G6PD TOMAH

G6PD, CYS385ARG
SNP: rs137852322, ClinVar: RCV000011101, RCV002305429

Hirono et al. (1989) demonstrated a T-to-C transition at nucleotide 1153, causing substitution of arginine for cysteine-385. The mutation created an Fnu4HI restriction site, which was used to confirm the mutation.


.0015   G6PD RIVERSIDE

G6PD, GLY410CYS
SNP: rs137852323, ClinVar: RCV000011102, RCV002305430

Hirono et al. (1989) demonstrated a G-to-T mutation at nucleotide 1228 that caused a change of glycine to cysteine at amino acid 410. The fact that the mutation destroyed an NciI restriction site was used to confirm the mutation.


.0016   G6PD MONTALBANO

G6PD, ARG285HIS
SNP: rs74575103, gnomAD: rs74575103, ClinVar: RCV000011080, RCV002288482, RCV003460450

Viglietto et al. (1990) found a new variant with nearly normal properties, due to a G-to-A transition that caused an arginine-to-histidine substitution at position 285. See Calabro et al. (1990).


.0017   G6PD RFLP

G6PD, NT1116, G-A
SNP: rs2230036, gnomAD: rs2230036, ClinVar: RCV000011083, RCV000079392, RCV001510602, RCV001540829, RCV002305751

D'Urso et al. (1988) found a silent G-to-A change at nucleotide 1116 (in exon 10), generating a PstI site.


.0018   G6PD RFLP

G6PD, NT1311, C-T
SNP: rs2230037, gnomAD: rs2230037, ClinVar: RCV000011084, RCV000079394, RCV001519331, RCV001719694, RCV002305751

De Vita et al. (1989) found a silent C-to-T change at nucleotide 1311 (in exon 11).


.0019   G6PD RFLP

G6PD, EX6, -60, C-G
SNP: rs2515904, gnomAD: rs2515904, ClinVar: RCV000011095, RCV001642221

Yoshida et al. (1988) found a RFLP resulting from a substitution in intron 5, creating a PvuII site. The probable change was C to G at a position 60 nucleotides upstream from exon 6 (Luzzatto, 1990).


.0020   G6PD ANDALUS

G6PD, ARG454HIS
SNP: rs137852324, gnomAD: rs137852324, ClinVar: RCV000011103, RCV002305431

Vives-Corrons et al. (1990) studied a G6PD variant resembling G6PD Mediterranean kinetically but with a slightly rapid electrophoretic mobility. They demonstrated a G-to-A transition at nucleotide 1361, producing an arg-to-his substitution.


.0021   G6PD CANTON

G6PD GIFU
G6PD AGRIGENTO
G6PD TAIWAN-HAKKA
G6PD, ARG459LEU
SNP: rs72554665, gnomAD: rs72554665, ClinVar: RCV000011104, RCV000011105, RCV000011106, RCV000011107, RCV000174272, RCV000375428, RCV000756195, RCV002305752, RCV002498469, RCV003460788

G6PD Canton is one of the most common deficient variants in Orientals, reaching a gene frequency of 1.7% in southern China (McCurdy et al., 1966). Stevens et al. (1990) demonstrated that codon 459 in G6PD-B is changed from CGT(arginine) to CTT(leucine). The G-to-T change occurs at nucleotide 1376. Tang et al. (1992) found this mutation in 3 Taiwanese and 1 Hakkanese in Taiwan. They pointed out that the same mutation occurs in 3 other Chinese G6PD variants in Guangdong, China: Taiwan-Hakka (McCurdy et al., 1970), Gifu (Fujii et al., 1984), and Agrigento (Sansone et al., 1975). The G6PD Gifu variant was discovered in a 9-year-old Japanese male with chronic hemolysis and hemolytic crises after upper respiratory infections (Fujii et al., 1984). Enzyme activity was 2.9% of normal. The patient's G6PD showed increased utilization of substrate analog, deamino-NADP, and thermal instability.


.0022   G6PD PUERTO LIMON

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, GLU398LYS
SNP: rs387906468, ClinVar: RCV000011108, RCV000066246, RCV003480029

Beutler et al. (1991) found a G-to-A transition at nucleotide 1192 causing a substitution of the amino acid lysine for the normal glutamic acid at position 398. This aberrant G6PD associated with nonspherocytic hemolytic anemia (300908) was described by Elizondo et al. (1982).


.0023   G6PD SANTAMARIA

G6PD, ASP181VAL, ASN126ASP
SNP: rs5030872, gnomAD: rs5030872, ClinVar: RCV000011073, RCV000011075, RCV000011109, RCV000030892, RCV000079405, RCV000178823, RCV000307631, RCV000477820, RCV000507037, RCV000761430, RCV000991016, RCV000999876, RCV001095678, RCV001267359, RCV001375611, RCV002305425, RCV002305432, RCV002305707, RCV002305727, RCV002305732, RCV002305803, RCV002305807, RCV002305808, RCV002305836, RCV002305857, RCV003460496

Beutler et al. (1991) found an A-to-T mutation at nucleotide 542 resulting in an asp-to-val substitution at amino acid 181. The subjects were white with 'some evidence of hemolysis' in one but none in the other. This aberrant G6PD, described by Saenz et al. (1984) in 2 unrelated subjects from Costa Rica, is 1 of 4 polymorphic variants that have 2 point mutations. One of these point mutations in each case is 376A-G (asn126asp), the change characteristic of the nondeficient polymorphic variant G6PD A+ (305900.0001).


.0024   G6PD GASTONIA

G6PD MARION
G6PD MINNESOTA
ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, VAL213LEU
SNP: rs137852326, gnomAD: rs137852326, ClinVar: RCV000011110, RCV000011111, RCV000011112, RCV000066251, RCV000790819, RCV002287328, RCV002305715

Beutler et al. (1991) found that although the patients from whom this variant G6PD was derived were unrelated, all had a G-to-T mutation at nucleotide 637 in exon 6 leading to substitution of leucine for valine-213. The G6PD variants called Gastonia, Marion, and Minnesota were all from patients with nonspherocytic hemolytic anemia (300908).


.0025   G6PD NASHVILLE

G6PD ANAHEIM
ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, ARG393HIS
ClinVar: RCV000011090, RCV000030890, RCV000030891, RCV000066231, RCV000756196, RCV003480028

In 2 unrelated patients with nonspherocytic hemolytic anemia (300908), Beutler et al. (1991) found a G-to-A mutation at nucleotide 1178 in exon 10 producing substitution of histidine for arginine-393.


.0026   G6PD VIANGCHAN

G6PD JAMMU
G6PD, VAL291MET
SNP: rs137852327, gnomAD: rs137852327, ClinVar: RCV000011116, RCV000011117, RCV000180545, RCV000405688, RCV000757320, RCV000763203, RCV001250220, RCV001266092, RCV002305706, RCV002305707, RCV003460452

See Poon et al. (1988). Beutler (1991) reported a G-to-A mutation at nucleotide 871, resulting in substitution of methionine for valine-291. The variant belonged to WHO class 2.

Louicharoen and Nuchprayoon (2005) and Matsuoka et al. (2005) indicated that G6PD Viangchan is the most common mutation in the Cambodian population, similar to Thai and Laotian populations, suggesting a common ancestry for people from these 3 countries. Matsuoka et al. (2005) found that G6PD Viangchan was linked in 8 cases with a 1311C-T transition (305900.0018) in exon 11 and a T-to-C substitution in intron 11, 93 bp downstream of exon 11. The finding was in accordance with studies of G6PD Viangchan in Laos, Thailand, and Malaysia.


.0027   G6PD A-

G6PD, ARG227LEU
SNP: rs137852328, gnomAD: rs137852328, ClinVar: RCV000011118, RCV002305857

In subjects with the G6PD A- phenotype, Hirono and Beutler (1988) found substitution of leucine for arginine-227, resulting from a G-to-T mutation at nucleotide 680 (rather than the val68-to-met mutation as in the usual G6PD A-). The mutation existed on the G6PD A background (asn126 to asp).


.0028   G6PD A-

G6PD, LEU323PRO
SNP: rs76723693, gnomAD: rs76723693, ClinVar: RCV000011119, RCV000757319, RCV000818410, RCV001375611, RCV003460453, RCV003934822

In subjects with the G6PD A- phenotype, Hirono and Beutler (1988) found substitution of proline for leucine-323, resulting from a T-to-C mutation at nucleotide 968 (rather than the val68-to-met mutation as in the usual G6PD A-). The mutation existed on the G6PD A background (asn126-to-asp).


.0029   G6PD KAIPING

G6PD ANANT
G6PD DHON
G6PD PETRICH-LIKE
G6PD SAPPORO-LIKE
G6PD, ARG463HIS
SNP: rs72554664, gnomAD: rs72554664, ClinVar: RCV000011120, RCV000011121, RCV000011122, RCV000011123, RCV000011124, RCV000174271, RCV000763201, RCV000823393, RCV000991190, RCV001266661, RCV003460789

Zuo et al. (1990) demonstrated substitution of histidine for arginine-463 resulting from a G-to-A mutation in nucleotide 1388. The G6PD was of the WHO class 2. The Chinese variant G6PD Kaiping was discovered by Du et al. (1988). The same mutation was found in G6PD Anant (Panich and Sungnate, 1973), Dhon (Panich and Na-Nakorn, 1980), Petrich-like (Shatskaya et al., 1980), and Sapporo-like (Fujii et al., 1981).


.0030   G6PD LOMA LINDA

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, ASN363LYS
SNP: rs137852329, ClinVar: RCV000011125, RCV000066262

In a patient with nonspherocytic hemolytic anemia (300908), Beutler et al. (1991) identified a C-to-A mutation at nucleotide 1089 in exon 10, producing substitution of asparagine-363 by lysine.


.0031   G6PD COIMBRA

G6PD, ARG198CYS
SNP: rs137852330, gnomAD: rs137852330, ClinVar: RCV000011126, RCV000011127, RCV000991015, RCV002305434, RCV003460454

In the son of a Portuguese woman who had suffered an attack of favism, Corcoran et al. (1992) identified a G6PD mutant with the chemical properties of the Mediterranean type (305900.0006). However, at the DNA level, they demonstrated that the mutation was a C-to-T transition 29 nucleotides downstream from the Mediterranean mutation, resulting in substitution of cysteine for arginine 10 amino acids downstream from the Mediterranean change. The same mutation was found in a patient in southern Italy. The new variant was called G6PD Coimbra.

In 3 individuals with G6PD deficiency from tribal groups in southern India, Chalvam et al. (2008) identified the Coimbra variant and stated that the mutation had a frequency of 7.5% in this population.


.0032   CHRONIC GRANULOMA AND HEMOLYTIC ANEMIA

G6PD, SER106CYS, ARG182TRP, ARG198CYS
SNP: rs267606835, rs267606836, gnomAD: rs267606835, rs267606836, ClinVar: RCV000011126, RCV000011127, RCV000991015, RCV002305434, RCV003460454

Gray et al. (1973) described a unique G6PD variant in a patient with chronic granuloma and hemolytic anemia. G6PD activity was undetectable not only in the patient's red blood cells but also in leukocytes and fibroblasts, and an immunologically crossreacting material was undetectable in these tissues. This is the only variant observed with no measurable activity and lack of crossreacting material, satisfying the definition for a 'null' variant. Maeda et al. (1992) found that the mRNA content and the size of mRNA were normal in the patient's lymphoblastoid cells (maintained as GM7254 in the Coriell repository in Camden, New Jersey). Western blot hybridization indicated that the patient's cells did not produce crossreacting material. Three nucleotide base changes were found in variant cDNA: a C-to-G transversion at nucleotide 317 (counting from adenine of the initiation codon), which should cause a ser-to-cys substitution at the 106th position (counting from the initiation met); a C-to-T transition at nucleotide 544, producing an arg-to-trp substitution at the 182nd position; and a C-to-T transition at nucleotide 592, resulting in an arg-to-cys substitution at the 198th position of the protein. No deletions or frameshift mutations were found, and no nucleotide change was detected in the extended 5-prime region which included the most distal cap site. When the variant cDNA was expressed in E. coli, the G6PD activity was about 2% of normal and crossreacting material was undetectable. However, when the variant mRNA was expressed in the in vitro translation system of rabbit reticulocytes, the variant protein was produced. The results suggested that extremely rapid in vivo degradation or precipitation of the variant enzyme induced by the 3 amino acid substitutions could be the major cause of the molecular deficiency.


.0033   G6PD TAIWAN-HAKKA 2

G6PD, ASN165ASP
SNP: rs137852331, gnomAD: rs137852331, ClinVar: RCV000011128, RCV000508226, RCV000723531, RCV000818047, RCV002504777, RCV003460455

Tang et al. (1992) identified an A-to-G transition at nucleotide 493 resulting in an asn165-to-asp amino acid substitution in the G6PD protein. The biochemical features of the mutation were not characterized. This mutation has only been reported in Chinese.

The Chinese population of Taiwan is divided into 4 groups: Taiwanese, mainland Chinese, Hakkanese, and Aborigines. The Taiwanese, the largest group, are descendants from emigrants who left mainland China during the 17th to 19th centuries. Most were from Fuchien Province on the southeast coast of China. The second largest population is mainland Chinese, who resided originally in many provinces throughout mainland China and migrated to Taiwan during the period 1948 to 1950. The third population is Hakkanese (Taiwan-Hakka), originally from Chung Yuan, who immigrated from the Kwangtung and Fuchien provinces on the southern coast of China and who came to Taiwan primarily during the 16th and 17th centuries. The native Taiwan Aborigines are a much smaller group, containing at least 9 distinct tribes whose ancestors are believed to have arrived in Taiwan from mainland Asia several thousand years ago. The frequency of G6PD deficiency varies from 4.52% in the Hakkanese to an average of 0.3% in most of the Aborigines. The Ami tribe of Aborigines shows a frequency of 3.5%, presumably a reflection of founder effect.


.0034   G6PD SANTIAGO

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, ARG198PRO
SNP: rs137852332, ClinVar: RCV000011129, RCV000011130

In a Chilean patient with nonspherocytic hemolytic anemia (300908), Beutler et al. (1992) identified a G-to-C transversion at nucleotide 593 leading to an arg198-to-pro substitution. They suggested G6PD Santiago as the designation. (G6PD Santiago de Cuba is a different mutation; see 305900.0009.)


.0035   G6PD MEXICO CITY

G6PD, ARG227GLN
SNP: rs137852328, gnomAD: rs137852328, ClinVar: RCV000011131, RCV001000765, RCV001168393, RCV001851785

In a Mexican individual with no clinical features attributable to the G6PD variant, Beutler et al. (1992) described a G-to-A transition at nucleotide 680 leading to an arg227-to-gln substitution. They suggested the designation G6PD Mexico City. (There is a G6PD Mexico; see under 305900.9999.) Nucleotide 680 is the same base that is altered from G-to-T in one type of G6PD A- (arg227-to-leu).


.0036   G6PD IERAPETRA

G6PD, PRO353SER
SNP: rs137852333, ClinVar: RCV000011115, RCV002305433, RCV003323355

In a Greek person with no clinical abnormalities that could be related to the G6PD variant, Beutler et al. (1992) identified a C-to-T transition at nucleotide 1057 resulting in a pro353-to-ser substitution.


.0037   G6PD GUADALAJARA

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, ARG387CYS
SNP: rs137852334, ClinVar: RCV000011132, RCV000011133, RCV002305727

See Vaca et al. (1982). In a Mexican patient with nonspherocytic hemolytic anemia (300908), Beutler et al. (1992) identified an arg387-to-cys substitution resulting from a C-to-T transition at nucleotide 1159.


.0038   G6PD ALHAMBRA

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, VAL394LEU
SNP: rs137852335, ClinVar: RCV000011134, RCV000011135

See Beutler and Rosen (1970). Beutler et al. (1992) indicated that the mutation in this G6PD variant found in a US white patient with nonspherocytic hemolytic anemia (300908) involved a G-to-C transversion at nucleotide 1180 resulting in a val394-to-leu substitution.


.0039   G6PD JAPAN

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, GLY410ASP
SNP: rs137852336, ClinVar: RCV000011136, RCV000011137

In a Japanese patient with nonspherocytic hemolytic anemia (300908), Beutler et al. (1992) identified a G-to-A transition at nucleotide 1229 resulting in a gly410-to-asp substitution.


.0040   G6PD PAWNEE

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, ARG439PRO
SNP: rs137852337, ClinVar: RCV000011138, RCV000011139, RCV000723442

In a US white patient with nonspherocytic hemolytic anemia (300908), Beutler et al. (1992) identified a G-to-C transition at nucleotide 1316 leading to an arg439-to-pro substitution.


.0041   G6PD SUNDERLAND

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, ILE35DEL
SNP: rs137852338, ClinVar: RCV000011140, RCV000011141, RCV001810842

Using a PCR-based technique, MacDonald et al. (1991) determined the nucleotide sequence of the entire coding region of the G6PD gene from a person with severe red cell G6PD deficiency and chronic hemolytic anemia (300908). The only abnormality found was a 3-bp deletion in exon 2, which predicted the loss of 1 of 2 adjacent isoleucine residues (amino acid 35 or 36), just upstream of the methionine residue called 'junctional' by Kanno et al. (1989). This part of exon 2 lies in a region that was thought by Kanno et al. (1989) to be encoded by a gene on chromosome 6, an idea subsequently disproved. The observations of MacDonald et al. (1991) demonstrated that a mutation in this X-linked amino-terminal region of G6PD caused deficiency in red cells. The deletion was within a 3-fold CAT repeat and had presumably resulted from misalignment at meiosis, with conservation of the reading frame.


.0042   G6PD KERALA-KALYAN

G6PD KERALA
G6PD KALYAN
G6PD, GLU317LYS
SNP: rs137852339, gnomAD: rs137852339, ClinVar: RCV000011142, RCV000011143, RCV000011144, RCV000800624, RCV001563664, RCV001815167, RCV003225020, RCV003466849

G6PD Kerala (Azevedo et al., 1968) and G6PD Kalyan (Ishwad and Naik, 1984), 2 variants discovered in India, were thought to be distinct on the basis of their biochemical properties. Ahluwalia et al. (1992) demonstrated that the molecular defect is identical. Both have a glu317-to-lys mutation which causes a loss of 2 negative charges; this is in keeping with the very slow electrophoretic mobility of G6PD Kerala-Kalyan. Both are accompanied by only mild enzyme deficiency. In both, the mutation is a C-to-T transition in the CpG dinucleotide. The mutations were found in 2 populations that are entirely distinct linguistically and culturally with no known historical links. However, in light of the traditional occupation of the Koli tribal group inhabiting the Kalyan district of Bombay, namely, marine fishing, victims of bad weather may have found their way to distant places where they were forced to live for some period, thus creating the possibility of gene flow.


.0043   G6PD AURES

G6PD, ILE48THR
SNP: rs76645461, gnomAD: rs76645461, ClinVar: RCV000011145, RCV000224890, RCV000704108, RCV001796957, RCV002305821, RCV002305849, RCV002482854, RCV003466850

In an Algerian boy who presented to the hospital with acute hemolytic anemia associated with 7 to 10% of G6PD residual activity, Nafa et al. (1993) identified a T-to-C transition at nucleotide 143 converting codon 48 from ATC (ile) to ACC (thr). The mutation was associated with favism.

In Saudi Arabia, Niazi et al. (1996) described G6PD Aures in 7 of 20 children (35%) with severe G6PD deficiency and in a 16-year-old boy with a history of passing dark urine after eating fava beans at the age of 5 years. Of the 20 children, 12 were positive for G6PD Mediterranean (305900.0006), and the mutation in 1 child remained unidentified.


.0044   G6PD GAOHE

G6PD, HIS32ARG
SNP: rs137852340, gnomAD: rs137852340, ClinVar: RCV000011146, RCV000798781, RCV002496323, RCV002512966, RCV003460456

This G6PD variant was described by Du et al. (1985). Its biochemical characterization was reviewed by Chiu et al. (1993), who demonstrated that the mutant is frequent in Chinese and consists of a change in cDNA nucleotide 95 from A to G (his to arg).


.0045   G6PD QUING YUAN

G6PD, GLY131VAL
SNP: rs137852341, gnomAD: rs137852341, ClinVar: RCV000011147, RCV000821638, RCV001810843, RCV002496324, RCV003460457

In an analysis of the molecular defect in 43 G6PD-deficient Chinese, Chiu et al. (1993) found 3 with a G-to-T transversion in cDNA nucleotide 392 (exon 5) resulting in a gly-to-val substitution. They reviewed the biochemical characteristics of this previously unidentified variant.


.0046   G6PD MAHIDOL-LIKE

G6PD, LEU342PHE
SNP: rs137852342, gnomAD: rs137852342, ClinVar: RCV000011148, RCV000079389, RCV000991013

In a study of the molecular defect in 43 G6PD-deficient Chinese, Chiu et al. (1993) identified a 'new' variant due to a C-to-T transition at cDNA nucleotide 1024 resulting in a leu-to-phe substitution. Chiu et al. (1993) listed the biochemical characteristics of G6PD Mahidol-like.


.0047   G6PD ORISSA

G6PD, ALA44GLY
SNP: rs78478128, gnomAD: rs78478128, ClinVar: RCV000011149, RCV000699943, RCV001538639, RCV003314551, RCV003466851

To determine the extent of heterogeneity of G6PD in India, Kaeda et al. (1995) studied several different Indian populations by screening for G6PD deficiency, followed by molecular analysis of deficient alleles. The frequency of G6PD deficiency varied between 3% and 15% in different tribal and urban groups. Remarkably, a previously unreported deficient variant, G6PD Orissa (ala44-to-gly), was found to be responsible for most of the G6PD deficiency in tribal Indian populations but was not found in urban populations where most of the G6PD deficiency was due to the G6PD Mediterranean (ser188-to-phe) variant (305900.0006). The distribution of G6PD alleles in India is reminiscent of the situation found with beta-globin (141900), as reviewed by Nagel and Ranney (1990). In that case, sickle cell anemia is almost entirely restricted to the tribal groups, whereas urban populations have a predominance of beta-thalassemia mutations. Kaeda et al. (1995) noted that the Km(NADP) of G6PD Orissa was 5-fold higher than that of the normal enzyme. This was thought to be due to the fact that the alanine residue that is replaced by glycine is part of a putative coenzyme-binding site. Surprisingly, the enzyme appeared to the authors to be more stable than normal G6PD, whereas most deficient variants have lowered stability.


.0048   G6PD NANKANG

G6PD, PHE173LEU
SNP: rs137852343, gnomAD: rs137852343, ClinVar: RCV000011150, RCV001060931, RCV003460458

In a Chinese newborn with neonatal jaundice, Chen et al. (1996) identified a novel G6PD mutation, G6PD NanKang, caused by a T-to-C transition at nucleotide 517, producing a phe173leu substitution in the G6PD protein.


.0049   G6PD MALAGA

G6PD, ASP181VAL
SNP: rs5030872, gnomAD: rs5030872, ClinVar: RCV000011109, RCV000030892, RCV000507037, RCV000991016, RCV002305432, RCV003460496

In a study of G6PD-deficient patients who presented with clinical favism in Spain, Vulliamy et al. (1996) found a new polymorphic variant they called G6PD Malaga, whose only abnormality was an A-to-T transversion at nucleotide 542 resulting in an asp181-to-val amino acid substitution. This was the same mutation previously found in association with the mutation of G6PD A-, namely asn126asp (305900.0001) in the double mutant G6PD Santamaria (305900.0023). G6PD Malaga was associated with enzyme deficiency class 3, and the enzymic properties of G6PD Malaga and G6PD Santamaria were quite similar. Vulliamy et al. (1996) speculated that G6PD Santamaria might have been produced by recombination between G6PDA and G6PD Malaga; however, haplotype analysis, including the use of a new silent polymorphism, suggested that the same 542A-T mutation had taken place independently in a G6PD B gene to give G6PD Malaga and in a G6PD A gene to give G6PD Santamaria.


.0050   G6PD NEAPOLIS

G6PD, PRO467ARG
SNP: rs137852344, gnomAD: rs137852344, ClinVar: RCV000011152, RCV002305435

In a study of 31 unrelated G6PD-deficient males in the Campania region of Southern Italy, Alfinito et al. (1997) found 9 different G6PD variants, 8 of which had already been described. The new variant, G6PD Neapolis, was found to have a pro467-to-arg substitution in the G6PD protein.


.0051   G6PD SERRES

ANEMIA, NONSPEHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, ALA361VAL
SNP: rs137852345, ClinVar: RCV000011153, RCV000143788

In a study of the causative mutation in 12 cases of G6PD deficiency associated with chronic nonspherocytic hemolytic anemia (300908), Vulliamy et al. (1998) found 1 patient to have a novel mutation, which they called G6PD Serres: a 1082C-T change, causing an ala361-to-val substitution in the dimer interface where most other severe G6PD mutations are found.


.0052   G6PD NARA

ANEMIA, NONSPEHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, 24-BP DEL, NT953
SNP: rs587776730, ClinVar: RCV000143789, RCV001509137

In a Japanese boy with severe G6PD deficiency (300908), Hirono et al. (1993) identified a 24-bp deletion (nucleotides 953-976) in exon 9 of the G6PD gene, which predicted an 8-amino acid deletion at residue 319.


.0053   G6PD AVEIRO

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, CYS269TYR
SNP: rs137852346, ClinVar: RCV000011155, RCV000011156

In a boy born in Aveiro, Portugal with severe chronic hemolytic anemia (300908) present from birth, Costa et al. (2000) found that the undetectable G6PD activity was caused by a G-to-A transition at nucleotide position 806 of the G6PD gene resulting in a cys269-to-tyr (C269Y) amino acid substitution. This mutation, which was designated G6PD Aveiro, was not detected in his mother or sister. By the age of 5 years, the patient had had 6 episodes of severe acute intravascular hemolysis that required hospitalization and erythrocyte transfusion. The spleen was palpable 6 cm below the left costal margin. Costa et al. (2000) pointed out that G6PD mutants causing class 1 variants (the most severe forms of the disease) cluster within exon 10, in a region that, at the protein level, is believed to be involved in dimerization. The mutation in this new class 1 variant maps to exon 8. Mutant and normal alleles were found in both hematopoietic and buccal cells, indicating mosaicism.


.0054   G6PD ASAHI

G6PD, VAL68MET
SNP: rs1050828, gnomAD: rs1050828, ClinVar: RCV000011075, RCV000011157, RCV000079404, RCV000224469, RCV000606259, RCV000999820, RCV001095679, RCV001267358, RCV002305425, RCV002305706, RCV002305732, RCV002305806, RCV002305807, RCV002305821, RCV003460495

G6PD A- is a common G6PD variant among Africans that may cause acute hemolysis triggered by infections and certain drugs, as well as by fava beans. This class 3 phenotype can be caused by a combination of the common 376A-G (asn126 to asp) mutation and either of 3 additional mutations that include 202G-A (val68 to met); see 305900.0002. The missense mutation 376A-G (asn126 to asp) by itself causes an asymptomatic class 4 variant G6PD A with normal enzyme activity, whereas the other mutation, 202G-A, had not been found in humans by itself. Hirono et al. (2002) described an asymptomatic G6PD-deficient patient with the missense mutation 202G-A but not the 376A-G. This was a 3-year-old Japanese boy who was noted to have jaundice and anemia on admission to the Asahi General Hospital. This was the only mutation found and it must have arisen separately from those common in Africans, because the patient had none of the silent mutations closely linked to the African mutation, while he had an intronic single base deletion common in Mongoloids. Town et al. (1992) had found in an in vitro study using recombinant human G6PD mutants expressed in E. coli that 202G-A, as well as 376A-G, does not cause enzyme deficiency by itself, and the synergistic action of these 2 mutations is necessary to produce the class 3 phenotype of G6PD A-. Synergistic interaction was also supported by the fact that val68 and asn126 are closely located in a 3-dimensional model of human G6PD. The results of Hirono et al. (2002) seem inconsistent with the idea that 202G-A cannot produce acute hemolysis by itself.


.0055   G6PD REHOVOT

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, TYR322HIS
SNP: rs137852347, ClinVar: RCV000011158, RCV000066276

In 3 brothers and their carrier mother of Jewish Ethiopian descent, Iancovici-Kidon et al. (2000) found a T-to-C transition at nucleotide 964 in exon 9 of the G6PD gene, resulting in a tyr322-to-his (Y322H) mutation. All 3 sibs showed hereditary nonspherocytic hemolytic anemia (300908), but the severity of hemolysis and the transfusion requirement varied markedly. One brother had severe congenital neutropenia (SCN; 202700), a condition not previously described in association with G6PD deficiency. Levels of white blood cell G6PD activity of the 3 sibs was 0 to 5% of normal controls. Neutrophil oxidative and bactericidal activities were impaired in the brother with SCN, but were well preserved in the other 2 sibs.


.0056   G6PD AMSTERDAM

ANEMIA, NONSPHEROCYTIC HEMOLYTIC, DUE TO G6PD DEFICIENCY
G6PD, 3-BP DEL, 180TCT
SNP: rs2070404412, ClinVar: RCV000066279

In a study of blood cells of 4 male patients from 2 unrelated families with nonspherocytic anemia (300908) and recurrent bacterial infections, van Bruggen et al. (2002) discovered that the activity of G6PD in red blood cells and in granulocytes was below detection level. Moreover, their granulocytes displayed a decreased respiratory burst upon activation. Sequencing of genomic DNA revealed a novel 3-bp (TCT) deletion in the G6PD gene, predicting the deletion of a leucine at position 61. The mutant G6PD protein was undetectable by Western blotting in red blood cells and granulocytes of these patients. In phytohemagglutinin-stimulated lymphocytes, the G6PD protein was present, but the amount of the protein was greatly diminished in the patients' cells. Purified mutant protein from an E. coli expression system showed decreased heat stability and decreased specific activity. Furthermore, van Bruggen et al. (2002) demonstrated that mRNA of the mutant G6PD was unstable, which may contribute to the severe G6PD deficiency observed in these patients. They proposed the name 'G6PD Amsterdam' for the new variant.

One family reported by van Bruggen et al. (2002) was Caucasian, the second was Hindustani. The Caucasian patient had an unremarkable medical history until he was admitted to the hospital at age 15 years with recurrent episodes of fever, jaundice, gastroenteritis, and coughing. He was found to have invasive disseminated aspergillosis (see 614079) in the lungs, brain, and soft tissues of the leg. Aspergillosis was successfully treated. Thereafter, hemoglobin level was normal but reticulocytosis was persistent. One of his brothers also had G6PD deficiency and presented with prolonged neonatal jaundice and episodes of acute hemolysis but had no known disposition to infections.

The Hindustani proband reported by van Bruggen et al. (2002) was healthy until the age of 3.5 years, when he was admitted to the hospital with pneumonia caused by Chromobacterium violaceum, an uncommon human pathogen that can cause serious infections in patients with neutrophil dysfunction. He was anemic. He responded well to chemotherapy, although the anemia persisted. With relapse he developed osteomyelitis but again responded to therapy.


.0057   G6PD ZURICH

G6PD, IVS10AS, A-G, -2
SNP: rs2070350038, ClinVar: RCV000011165, RCV002305437

In a 33-year-old Swiss male with G6PD deficiency, designated G6PD Zurich, Efferth et al. (2004) identified a single nucleotide mutation that altered position -2 of intron 10 of the G6PD gene from the consensus A to G. The mutation resulted in alternative splicing that removed the first 9 nucleotides of exon 11, which code for amino acids asparagine, valine, and lysine at positions 430-432, respectively. Efferth et al. (2004) estimated that 400 million people worldwide are affected by G6PD deficiency, the most common hereditary enzymopathy, with some 140 known molecular G6PD defects. They pointed out that most mutations in the G6PD gene are missense mutations. To the best of their knowledge, there was only 1 missplicing mutation previously described: G6PD Varnsdorf (305900.0058), which is caused by destruction of the same obligate splice site as that destroyed in G6PD Zurich. In G6PD Varnsdorf, the invariant 3-prime AG dinucleotide has been deleted, whereas in G6PD Zurich, a point mutation has changed AG to GG. In both G6PD Zurich and G6PD Varnsdorf, the next available downstream consensus splice sequence is used, resulting in deletion of 3 amino acids. Efferth et al. (2004) suggested that it was no coincidence that the only 2 splicing mutations of G6PD identified to that time both affected the same splice site. Since null mutations of G6PD appear to be incompatible with life, a functional alternative splice site that does not cause a frameshift is required for viability. The 3-prime splice site of intron 10 offers this opportunity.


.0058   G6PD VARNSDORF

G6PD, IVS10AS, 2-BP DEL, AG, -2
SNP: rs2070350009, ClinVar: RCV000011166, RCV002305438

See 305900.0057 and Efferth et al. (2004).


.0059   G6PD COSENZA

G6PD, ARG459PRO
SNP: rs72554665, gnomAD: rs72554665, ClinVar: RCV000011167, RCV000354491, RCV000778152, RCV001212765, RCV003460459

Calabro et al. (1993) identified a novel G6PD variant, which they called Cosenza, in patients with G6PD deficiency from the Calabria region of southern Italy. The arg459-to-pro (A459P) substitution results from a 1376G-C transversion. The mutant protein retains less than 10% enzyme activity and belongs to the group of severe disorders often associated with hemolysis.

Barisic et al. (2005) identified G6PD Cosenza in 9 (37.5%) of 24 unrelated patients with G6PD deficiency from the Dalmatian region of southern Croatia. Seven of the 9 patients had favism.


.0060   G6PD SPLIT

G6PD, PRO481ARG
SNP: rs137852348, ClinVar: RCV000011159, RCV002305436

In a male with G6PD deficiency from the Dalmatian region of southern Croatia, Barisic et al. (2005) identified a 1442C-G transversion in the G6PD gene, resulting in a pro481-to-arg (P481R) substitution. The mutant protein retained approximately 30% enzyme activity (class 3).


.0061   G6PD NAMORU

G6PD, HIS70TYR
SNP: rs137852349, gnomAD: rs137852349, ClinVar: RCV000011160, RCV001264782

Chalvam et al. (2007) identified a 208T-C transition in exon 4 of the G6PD gene, resulting in a his70-to-tyr (H70Y) substitution, as the basis of G6PD deficiency in Indian patients with the disorder. The H70Y mutation was detected in 28 (70.4%) of 40 affected Indian males from 3 tribal groups from the Nilgiri district of Tamil Nadu in southern India. The variant was termed G6PD Namoru.


.0062   G6PD NILGIRI

G6PD, ARG198HIS
SNP: rs137852332, ClinVar: RCV000011161, RCV000991014

In 4 individuals with G6PD deficiency from tribal groups of the Nilgiri district in southern India, Chalvam et al. (2008) identified a 593G-A transition in exon 6 of the G6PD gene, resulting in an arg198-to-his (R198H) substitution, which they designated G6PD Nilgiri. The authors stated that the mutation had a frequency of 10.0% in this population.


.9999   GLUCOSE-6-PHOSPHATE DEHYDROGENASE VARIANTS, MOLECULAR DEFECT UNKNOWN

G6PD VARIANTS, MOLECULAR DEFECT UNKNOWN

The following list of G6PD variants which have not been characterized at the molecular level is in alphabetic order. Quotation marks surround the name of each G6PD variant about which there is insufficient information for certainty of its uniqueness.

G6PD AACHEN. See Kahn et al. (1976).

G6PD AARAU. See Gahr et al. (1976).

G6PD 'ABEOKUTA'. See Usanga et al. (1977).

G6PD ABRAMI. See Kahn et al. (1975).

G6PD 'ADAME'. See Usanga et al. (1977).

G6PD ADANA. See Aksoy et al. (1987).

G6PD AKITA. See Miwa et al. (1978).

G6PD ALABAMA. Prchal et al. (1988) described a 6-year-old black boy who had transient hemolysis after a viral infection and was found to have mildly decreased red cell G6PD activity. The unusual finding was the presence of 2 G6PD bands in him and in his maternal grandfather despite normal XY karyotype. Two bands were seen only in reticulocytes. Prchal et al. (1988) postulated that there were 2 transcriptional products of the mutant G6PD gene, 1 of which had a short half-life and was detectable only in young red blood cells.

G6PD ALBUQUERQUE. See Beutler et al. (1968).

G6PD ALESSANDRIA. Similar to G6PD Alexandra. See Sansone et al. (1981).

G6PD ALEXANDRA. This was found in Australia in a male of Italian extraction who suffered severe neonatal jaundice following maternal ingestion of fava beans prenatally and postnatally. Retesting in adolescence showed milder expression of the enzyme defect (Harley et al., 1978).

G6PD ALGER. See Benabadji et al. (1978).

G6PD AMBOIN. See Chockkalingam et al. (1982).

G6PD AMMAN-1. See Karadsheh et al. (1986).

G6PD AMMAN-2. See Karadsheh et al. (1986).

G6PD ANGORAM. See Chockkalingam et al. (1982).

G6PD ANKARA. See Kahn et al. (1975).

G6PD ARLINGTON HEIGHTS. See Honig et al. (1979).

G6PD ASAHIKAWA. This variant was discovered in a 6-year-old Japanese boy with chronic hemolytic anemia and hemolytic crises after upper respiratory infections (Takizawa et al., 1984).

G6PD ASHDOD. See Ramot et al. (1969).

G6PD ATHENS. See Stamatoyannopoulos et al. (1967).

G6PD 'ATHENS-LIKE'. See Stamatoyannopoulos et al. (1971).

G6PD ATLANTA. See Beutler et al. (1976).

G6PD 'ATTICA'. See Rattazzi et al. (1969).

G6PD AVENCHES. See Pekrun et al. (1989).

G6PD 'AVVOCATA'. See Colonna-Romano et al. (1985).

G6PD AYUTTHAYA. See Panich (1980).

G6PD AZERBAIJAN. See Shatskaya et al. (1975).

G6PD B. The so-called normal, this form predominates in all populations greater than a few hundred (Yoshida et al., 1971).

G6PD 'BAGDAD'. See Geerdink et al. (1973).

G6PD BAKU. See Shatskaya et al. (1980).

G6PD 'BALCALI'. See Aksoy et al. (1987).

G6PD BALI. See Chockkalingam et al. (1982).

G6PD BALTIMORE-AUSTIN. See Porter et al. (1964) and Long et al. (1965).

G6PD BANGKOK. See Talalak and Beutler (1969).

G6PD BARBIERI. See Marks et al. (1962).

G6PD BARCELONA. See Vives-Corrons et al. (1982). This is one of the rare G6PD variants associated with granulocyte dysfunction and increased susceptibility to infections. Hemolysis in this form of chronic nonspherocytic hemolytic anemia is exaggerated by infection.

G6PD 'BASH-KUNGUT I AND II'. See Shatskaya et al. (1980).

G6PD 'BASH-KUNGUT IV'. See Shatskaya et al. (1980).

G6PD BAT-YAM. See Ramot et al. (1969).

G6PD BAUDELOCQUE. See Junien et al. (1974).

G6PD 'BEAUJON'. See Boivin and Galand (1968).

G6PD BEAUMONT. Mamlok et al. (1985) reported a new molecular variant associated with severe enzyme deficiency and chronic nonspherocytic hemolytic anemia. The characteristics were marked heat lability, a normal rate constant value for glucose-6-phosphate, a nearly normal pH activity curve, and increased use of 2-deoxyglucose-6-phosphate. Mamlok et al. (1987) described a fatal case of Chromobacterium violaceum sepsis in a 3-year-old boy with this variant. The child was an identical twin; the surviving twin subsequently had a severe episode of Campylobacter jejuni gastroenteritis. Patients with severe deficiency of G6PD and polymorphonuclear leukocytes have increased susceptibility to infections and abnormal phagocyte function that resembles that of patients with chronic granulomatous disease, but such had not hitherto been reported during the first decade of life. Infections with C. violaceum are rare; most of the 20 or so infections have occurred in Louisiana or Florida and have been associated with warm, stagnant water sources.

G6PD BENEVENTO. See McCurdy et al. (1973).

G6PD BERLIN. See Helge and Borner (1966).

G6PD BIDEIZ. See Krasnopolskaya et al. (1977).

G6PD BIELEFELD. See Gahr et al. (1977).

G6PD BIRMINGHAM. See Prchal et al. (1980).

G6PD BLIDA. See Benabadji et al. (1978).

G6PD BNEI BRAK. See Sidi et al. (1980).

G6PD BODENSEE. See Benohr et al. (1971).

G6PD BOGIA. See Chockkalingam and Board (1980).

G6PD BOLUO. See Du et al. (1988).

G6PD BOLUO-2. See Du et al. (1988).

G6PD BOSTON. See Necheles et al. (1971).

G6PD BUKITU. See Chockkalingam and Board (1980).

G6PD BUTANTAN. In Brazil, Stocco dos Santos et al. (1991) described a Gd(+) variant which was characterized by normal activity and electrophoretic mobility, increased Km, and increased activity for 2-deoxy-G6P. The variant, which they called G6PD Butantan, was present in 3, and perhaps a fourth, cousin; the 4 mothers were sisters. All 4 males had severe mental retardation, bilateral congenital hip luxation, and short stature. Five uncles of these males may have been affected. In this family, Stocco dos Santos et al. (2003) found linkage of the X-linked mental retardation syndrome (300434) to the pericentric region, Xp11.3-q21.1.

G6PD 'CAGLIARI II' (CAGLIARI-LIKE). See Frigerio et al. (1987) and Calabro et al. (1990).

G6PD 'CALTANISSETTA'. See Sansone et al. (1981) and Perroni et al. (1982).

G6PD 'CAMALDOLI'. See Colonna-Romano et al. (1985).

G6PD CAMPBELLPORE. See McCurdy et al. (1970).

G6PD CAMPERDOWN. Harley et al. (1978) found this variant in Australia in a boy of Maltese extraction in whom lamellar cataracts were found at age 4. The enzyme deficiency was detected in a screening of children of Mediterranean extraction with lamellar cataracts. The boy had no excessive hemolysis. Previous descriptions of cataracts were in patients with hemolytic anemia.

G6PD CAPETOWN. See Botha et al. (1969).

G6PD CARSWELL. See Siegel and Beutler (1971).

G6PD CASTILLA-LIKE. See Chockkalingam et al. (1982).

G6PD CAUJERI. See Gutierrez et al. (1987).

G6PD CENTRAL CITY. See Csepreghy et al. (1988).

G6PD CHAINAT. See Panich and Na-Nakorn (1980).

G6PD CHAO PHYA. See Panich (1980).

G6PD CHARLESTON. See Beutler et al. (1972).

G6PD CHIAPAS. See Lisker et al. (1978).

G6PD CHIBUTO. See Reys et al. (1970).

G6PD CHICAGO. See Kirkman et al. (1964) and Fairbanks et al. (1980). Fairbanks et al. (1980) demonstrated that G6PD Chicago and G6PD Cornell are the same variant; they had been described previously in different members of a single large kindred.

G6PD CHINESE. See Chan et al. (1972).

G6PD CIUDAD DE LA HABANA. See Gonzalez et al. (1980).

G6PD 'CLICHY'. See Boivin and Galand (1968).

G6PD CLINIC. In a young patient with chronic nonspherocytic hemolytic anemia and familial amyloidotic polyneuropathy, Vives-Corrons et al. (1989) identified a new variant with a markedly acidic pH optimum. It bore some similarity in its molecular characteristics to G6PD Bangkok and G6PD Duarte.

G6PD COLOMIERS. See Vergnes et al. (1981).

G6PD COLUMBUS. See Pinto et al. (1966).

G6PD CORINTH. Yoshida, A.: unpublished, 1975.

G6PD CORNELL. See Miller and Wollman (1974) and Fairbanks et al. (1980). Fairbanks et al. (1980) demonstrated that G6PD Chicago and G6PD Cornell are the same variant; they had been described previously in different members of a single large kindred.

G6PD CUIABA. In a 33-year-old male of Portuguese extraction who developed hemolytic anemia after acetaminophen and acetylsalicylic acid ingestion, Barretto and Nonoyama (1987) found a variant G6PD which had normal activity and normal electrophoretic mobility, but unusually high K(m) for glucose-6-phosphate, high K(i) for NADPH, and decreased thermal stability.

G6PD 'DAKAR'. See Kahn et al. (1971, 1973).

G6PD DALLAS. Beutler, E.; Frenkel, E. P.; Forman, L.: unpublished, 1987.

G6PD DEBROUSSE (G6PD CONSTANTINE, FORMERLY). See Kissin and Cotte (1970) and Sansone et al. (1975).

G6PD DJYNET. See Krasnopolskaya and Bochkov (1982).

G6PD DOTHAN. See Prchal et al. (1979).

G6PD DUARTE. See Beutler et al. (1968).

G6PD DUBLIN. See McCann et al. (1980).

G6PD DUSHANBA I. See Krasnopolskaya and Bochkov (1982).

G6PD DUSHANBA II. See Krasnopolskaya and Bochkov (1982).

G6PD DUSHANBA III. See Krasnopolskaya and Bochkov (1982).

G6PD EAST AFRICAN. See Othieno-Obel (1972).

G6PD EAST HARLEM. See Feldman et al. (1977).

G6PD 'EKITI'. See Usanga et al. (1977).

G6PD EL-FAYOUM. See McCurdy et al. (1974).

G6PD EL-KHARGA. See McCurdy et al. (1974).

G6PD EL MORRO. See McCurdy et al. (1973).

G6PD ENGLEWOOD. See Rattazzi et al. (1971).

G6PD 'ENSLEY'. See Nsouly and Prchal (1981).

G6PD 'ESPOO'. See Vuopio et al. (1975).

G6PD FERRANDINA. See Calabro et al. (1990).

G6PD FERRARA. See Carandina et al. (1976).

G6PD FERRARA II. See De Flora et al. (1981) and Sansone et al. (1981).

G6PD 'FERRARA III'. See Perroni et al. (1982).

G6PD FORT PIERCE. Phyliky, R. L.; Nishimura, R. A. and Beutler, E.: unpublished, 1983.

G6PD FORT WORTH. See Mills et al. (1975).

G6PD 'FRANKFURT'. Nowicki et al. (1974).

G6PD FREIBURG. See Weinreich et al. (1968) and Busch and Boie (1970).

G6PD FUKUOKA. This variant was found in a 77-year-old male with drug-induced hemolysis (Fujii et al., 1984). Enzyme activity was 6.4% of normal and the patient's G6PD had abnormal electrophoretic mobility and thermal instability.

G6PD FUKUSHIMA. Miwa et al. (1978) described this 'deficiency' mutant, which leads to chronic hemolytic anemia. It was slow-moving electrophoretically, like G6PD Kurume, from which it differed by low utilization of deamino-NADP and normal pH curve. The proband, a 33-year-old male, had 2.8% of normal enzyme activity and mild hemolytic anemia. Miwa et al. (1978) stated that 46 variants had previously been classified as class 1, with severe enzyme deficiency leading to chronic nonspherocytic hemolytic anemia.

G6PD GABROVIZZA. See Ventura et al. (1984).

G6PD 'GALLIERA'. See Perroni et al. (1982).

G6PD GALLURA. See Sansone et al. (1975).

G6PD 'GALVESTON'. See Alperin and Mills (1972).

G6PD 'GAMBIA'. Welch et al. (1978) found a gene frequency of 0.024 among 1,109 persons examined in The Gambia. This is a slow electrophoretic variant with reduced enzyme activity.

G6PD GAOMIN. See Du et al. (1988).

G6PD GAOZHOU. See Du et al. (1988).

G6PD GENOVA. See Gaetani et al. (1990).

G6PD GOODENOUGH. See Chockkalingam et al. (1982).

G6PD GOTZE DELCHEV. See Shatskaya et al. (1980). G6PD GRAND PRAIRIE. See Cederbaum and Beutler (1975).

G6PD GREAT LAKES. Beutler, E. and Maurer, H. S.: unpublished, 1984.

G6PD GUANGZHOU. See Du et al. (1988).

G6PD GUANTANAMO. See Gutierrez et al. (1987). G6PD 'GUIBA'. See Weimer et al. (1981).

G6PD HAAD YAI. See Panich and Na-Nakorn (1980).

G6PD 'HAMBURG'. See Gahr and Schroter (1974).

G6PD HAMM. See Gahr et al. (1976).

G6PD 'HANOI'. See Toncheva (1986).

G6PD HAWAII. Beutler, E. and Matsumoto, F.: unpublished, 1975.

G6PD HAYEM. See Kahn et al. (1974). G6PD HEIAN. See Nakai and Yoshida (1974).

G6PD HEKTOEN. Substitution of tyrosine for histidine (Dern et al., 1969).

G6PD HELSINKI. See Vuopio et al. (1973) and Harkonen and Vuopio (1974). Cohn et al. (1979) described severe hemolytic anemia in 2 Danish boys, who showed deficiency of G6PD. The enzyme had characteristics possibly identical to those of G6PD Helsinki.

G6PD HILLBROW. See Cayanis et al. (1975).

G6PD 'HIROSHIMA-1'. See Kageoka et al. (1985).

G6PD 'HIROSHIMA-2'. See Kageoka et al. (1985).

G6PD 'HIROSHIMA-3'. See Kageoka et al. (1985).

G6PD HOFU. See Miwa et al. (1977).

G6PD HONG KONG. See Wong et al. (1965) and Chan et al. (1972).

G6PD HONG KONG POKFULAM. See Chan et al. (1972).

G6PD HOTEL DIEU. See Kahn et al. (1977).

G6PD HUALIEN. McCurdy, P. R.: unpublished, 1975.

G6PD HUALIEN-CHI. McCurdy, P. R.: unpublished, 1975.

G6PD HUAZHOU. See Du et al. (1988).

G6PD HUIYANG. See Du et al. (1988).

G6PD HUNTSVILLE. See Hall et al. (1988).

G6PD HURON. See Ravindranath and Beutler (1987).

G6PD IBADAN-AUSTIN. See Long et al. (1965).

G6PD IJEBU-ODE. See Luzzatto and Afolayan (1968).

G6PD INDIANAPOLIS. Beutler, E.; Forman, L.; Gelbart, T.: unpublished, 1985.

G6PD INDONESIA. See Kirkman and Eng (1969).

G6PD INHAMBANE. See Reys et al. (1970).

G6PD INTANON. See Panich (1974).

G6PD ISERLOHN. Unstable enzyme. See Eber et al. (1985).

G6PD ITA-BALE. See Long et al. (1965).

G6PD IWATE. See Kanno et al. (1987).

G6PD JACKSON. See Thigpen et al. (1974).

G6PD JALISCO. See Vaca et al. (1985).

G6PD JOHANNESBURG. See Balinsky et al. (1973).

G6PD 'JUNUT'. See Shatskaya et al. (1980).

G6PD KABYLE. See Kaplan et al. (1967).

G6PD KALUAN. See Chockkalingam and Board (1980).

G6PD KALUGA. See Shatskaya et al. (1976).

G6PD KAMIUBE. See Nakatsuji and Miwa (1979).

G6PD KAN. See Panich (1973).

G6PD KANAZAWA. This variant, found by Kitao et al. (1982) in a Japanese male with chronic nonspherocytic hemolytic anemia, has normal electrophoretic mobility, normal Km for glucose-6-phosphate and NADP, and normal utilization of the substrate 2-deoxyglucose-6-phosphate and deamino-NADP. It shows decreased thermal stability and a biphasic pH curve. G6PD KAR KAR. See Chockkalingam et al. (1982).

G6PD KARDISTA. Stamatoyannopoulos, G.: unpublished, 1975.

G6PD KEPHALONIA. See Rattazzi et al. (1969).

G6PD KEROVOGRAD. See Krasnopolskaya and Bochkov (1982). G6PD 'KHARTOUM'. See Samuel et al. (1981).

G6PD 'KILGORE'. See Alperin and Mills (1972).

G6PD KING COUNTY. Yoshida, A.: unpublished, 1975.

G6PD KIROVOGRAD. See Shatskaya et al. (1976).

G6PD KIWA. See Nakatsuji and Miwa (1979).

G6PD KOBE. See Fujii et al. (1981).

G6PD KONAN. See Nakatsuji and Miwa (1979).

G6PD KREMENCHUNG. See Cherniak et al. (1977) and Tokarev et al. (1978).

G6PD KUANYAMA. See Balinsky et al. (1974).

G6PD KURUME. A 'deficiency' mutation, this variant leads to chronic hemolytic anemia. It is electrophoretically slow-moving. The proband was a 17-year-old male whose red cells had only 0.8% normal enzyme activity (Miwa et al., 1978). The enzyme showed normal KmG6P, normal KmNADP, low KiNADP, normal utilization of 2-deoxy-G6P and deamino-NADP, very low heat stability, and a biphasic pH curve.

G6PD 'KYOTO'. See Kojima (1972). G6PD LAGHOUAT. See Benabadji et al. (1978).

G6PD LAGUNA. Although the proband was anemic, the absence of anemia in relatives with the same G6PD variant suggested that the association was coincidental (Weimer et al., 1984). The characteristics of the mutant enzyme, including slower electrophoretic mobility, were described.

G6PD 'LANLATE'. See Usanga et al. (1977).

G6PD LAOS. Smith, J. W. and Beutler, E.: unpublished, 1981.

G6PD LAWNDALE. See Grossman et al. (1966).

G6PD LEVADIA. See Stamatoyannopoulos et al. (1970).

G6PD LIFTA. See Ramot et al. (1969).

G6PD LINCOLN PARK. See Honig et al. (1979).

G6PD LINDA VISTA. Smith, J. W. and Beutler, E.: unpublished, 1981.

G6PD 'LIZU-BAISHA'. See Du (1981).

G6PD LONG PRAIRIE. See Johnson et al. (1977).

G6PD LONG XUYEN. See Panich et al. (1980).

G6PD LOS ANGELES. See Beutler and Matsumoto (1977).

G6PD LOURENZO MARQUES. See Reys et al. (1970). G6PD LOZERE. See Vergnes et al. (1976).

G6PD LUBLIN. See Pawlak et al. (1970).

G6PD LUZ-SAINT SAUVEUR. See Vergnes et al. (1973).

G6PD LYNN (G6PD YUGOSLAVIA, FORMERLY). Beutler, E. and Lind, S.: unpublished, 1987.

G6PD MADANG. See Chockkalingam et al. (1982).

G6PD MADISON. See Shows et al. (1964).

G6PD MADRONA. See Hook et al. (1968).

G6PD MAINOKI. See Chockkalingam et al. (1982).

G6PD 'MALI'. See Kahn et al. (1971).

G6PD MAMMOLA. See Perroni et al. (1982).

G6PD MANCHESTER. See Milner et al. (1974).

G6PD MANDANG. See Chockkalingam et al. (1982).

G6PD MANJACAZE. See Reys et al. (1970).

G6PD MANUS. See Chockkalingam et al. (1982).

G6PD MARKHAM. See Kirkman et al. (1968).

G6PD 'MARTINIQUE'. See Kahn et al. (1971).

G6PD MARTINIQUE-LIKE. See Krasnopolskaya et al. (1977).

G6PD MATAM. See Kahn et al. (1975).

G6PD MELISSA. Stamatoyannopoulos, G.: unpublished, 1975.

G6PD MENORCA. See Vives-Corrons and Pujades (1982).

G6PD MERCURY. Beutler, E. and Taylor, G. P.: unpublished, 1982.

G6PD MEXICO. See Lisker et al. (1972).

G6PD MIAOZU-BAISHA. See Du et al. (1984).

G6PD MILWAUKEE. See Westring and Pisciotta (1966).

G6PD MINAS GERAIS. See Azevedo and Yoshida (1969).

G6PD MINNEAPOLIS. Johnson, G. J. and Beutler, E.: unpublished, 1980.

G6PD 'MISENO'. See Colonna-Romano et al. (1985).

G6PD MISSOULA. See Wilson (1976).

G6PD MOOSBURG. See Pekrun et al. (1989).

G6PD MORELIA. Class 4. First in class with a high Km for NADP and a low Ki for NADPH. See Vaca et al. (1985).

G6PD MOSCOW. See Batischev et al. (1977).

G6PD MURET. See Vergnes et al. (1981).

G6PD MUSASHINO. See Kumakawa et al. (1987).

G6PD NAGANO. This variant is associated with infection-induced hemolysis and chronic hemolytic anemia due to markedly impaired enzyme activity and thermal instability (Takahashi et al., 1982).

G6PD 'NAGASAKI-1'. See Kageoka et al. (1985).

G6PD 'NAGASAKI-2'. See Kageoka et al. (1985).

G6PD 'NAGASAKI-3'. See Kageoka et al. (1985).

G6PD 'NANCY'. See Streiff and Vigneron (1971).

G6PD NANHAI. See Du et al. (1988).

G6PD NAPOLI. See De Flora et al. (1981).

G6PD NEDELINO. See Toncheva and Tzoneva (1984).

G6PD NEW GUINEA-II. See Rattazzi et al. (1971).

G6PD NEW YORK. See Rattazzi et al. (1971).

G6PD N-PATHOM. See Panich (1974) and Panich and Na-Nakorn (1980).

G6PD N-SAWAN. See Panich and Na-Nakorn (1980).

G6PD NUCUS. See Yermakov et al. (1981).

G6PD NUHA. See Krasnopolskaya and Bochkov (1982).

G6PD 'NUKHA'. See Shatskaya et al. (1980).

G6PD OGIKUBO. See Miwa et al. (1978).

G6PD OGORI. See Lisker et al. (1977). G6PD OHIO. See Pinto et al. (1966).

G6PD OKHUT I. See Krasnopolskaya et al. (1977).

G6PD OKHUT II. See Krasnopolskaya et al. (1977).

G6PD OKLAHOMA. See Kirkman and Riley (1961) and Nance (1964).

G6PD ONODA. Nakashima, K.: unpublished, 1978.

G6PD ORCHOMENOS. See Stamatoyannopoulos et al. (1971).

G6PD PADREW. See Panich and Na-Nakorn (1980).

G6PD PALAKAU. See Chockkalingam et al. (1982).

G6PD 'PALEPOLI'. See Colonna-Romano et al. (1985).

G6PD 'PALLONETTO'. See Colonna-Romano et al. (1985).

G6PD 'PALMI I'. See Perroni et al. (1982).

G6PD 'PALMI II'. See Perroni et al. (1982).

G6PD PANAMA. See Beutler et al. (1974).

G6PD PANAY. See Fernandez and Fairbanks (1968).

G6PD PANAY-LIKE.

G6PD 'PARIS'. See Boivin and Galand (1968).

G6PD PEA RIDGE. See Fairbanks et al. (1980).

G6PD 'PETILIA'. See Sansone et al. (1981) and Perroni et al. (1982).

G6PD PETRICH. See Shatskaya et al. (1980).

G6PD PINAR DEL RIO. See Gonzalez et al. (1977).

G6PD PISTICCI. See Viglietto et al. (1990) and Calabro et al. (1990).

G6PD POMPTON PLAINS. Beutler, E.; Davis, S.; Forman, L. and Gelbart, T.: unpublished, 1985.

G6PD POPONDETTA. See Chockkalingam et al. (1982).

G6PD PORBANDAR. See Cayanis et al. (1977).

G6PD 'PORDENONE'. See Sansone et al. (1981) and Perroni et al. (1982).

G6PD PORT ELIZABETH. See Balinsky et al. (1973).

G6PD PORT-ROYAL. See Kaplan et al. (1971).

G6PD PORTO ALEGRE. See Hutz et al. (1977).

G6PD 'POSILIPPO'. See Colonna-Romano et al. (1985).

G6PD POZNAN. See Pawlak et al. (1975).

G6PD 'POZZALLO'. See Perroni et al. (1982).

G6PD PUERTO RICO. See McCurdy et al. (1973).

G6PD QING-BAILJIANG. See Du et al. (1988).

G6PD RAMAT-GAN. See Ramot et al. (1969).

G6PD REGAR. See Ermakov et al. (1983).

G6PD REGENSBURG. See Eber et al. (1985).

G6PD 'RENNES'. See Picat et al. (1980).

G6PD ROTTERDAM. See Rattazzi et al. (1971).

G6PD RUDOSEM. See Toncheva and Tzoneva (1984).

G6PD RUSSIAN-MOSCOW. See Krasnopolskaya and Bochkov (1982).

G6PD SALATA. See Chockkalingam and Board (1980).

G6PD SAMANDAG. See Aksoy et al. (1987).

G6PD SAN DIEGO. See Howell et al. (1972).

G6PD SAN FRANCISCO. See Mentzer et al. (1980).

G6PD SAN JOSE. See Castro and Snyder (1974).

G6PD SAN JUAN. See McCurdy et al. (1973).

G6PD SANTA BARBARA. Kidder, W. R. and Beutler, E.: unpublished, 1979.

G6PD SAPPORO. See Fujii et al. (1981).

G6PD 'SCHWABEN'. See Benohr et al. (1971).

G6PD 'S.DONA'. See Perroni et al. (1982).

G6PD SEATTLE. See Kirkman et al. (1965).

G6PD SELIM. See Shatskaya et al. (1975).

G6PD SENDAGI. This variant was associated with chronic nonspherocytic hemolytic anemia in a 2-year-old Japanese male in whom upper respiratory infection precipitated a hemolytic crisis (Morisaki et al.,1983).

G6PD SHEKII. See Krasnopolskaya et al. (1977).

G6PD SHIRIN-BULAKH. See Krasnopolskaya et al. (1977).

G6PD SIRIRAJ. See Panich et al. (1972).

G6PD SIWA. See McCurdy et al. (1974).

G6PD SONGKHLA. See Panich and Na-Nakorn (1980).

G6PD S-SAKORN. See Panich (1980).

G6PD ST. LOUIS. See Kahn et al. (1974).

G6PD STEILACOM. Yoshida, A.; Baur, E. and Voigtlander, B.: unpublished, 1975.

G6PD 'STELLA'. See Colonna-Romano et al. (1985).

G6PD 'STRASBOURG'. See Waitz et al. (1970).

G6PD SWIT. See Chockkalingam et al. (1982).

G6PD TACOMA. Yoshida, A. and Baur, E.: unpublished, 1975.

G6PD TACOMA-LIKE. See Vergnes et al. (1975).

G6PD TAHTA. See McCurdy et al. (1974).

G6PD TAIPEI-HAKKA. See McCurdy et al. (1970).

G6PD 'TAIWAN-AMI 5'. See McCurdy et al. (1970).

G6PD 'TAIWAN-AMI 6'. See McCurdy et al. (1970).

G6PD TARSUS. See Gahr et al. (1976).

G6PD TASHKENT. See Yermakov et al. (1981).

G6PD TEHERAN. McCurdy, P. R.: unpublished, 1965.

G6PD TEL HASHOMER. See Ramot and Brok (1964) and Kirkman et al. (1969).

G6PD TENGANAN. See Chockkalingam et al. (1982).

G6PD THENIA. See Benabadji et al. (1978).

G6PD THESSALONIKI. Koliakos et al. (1989) found a new variant in a 70-year-old patient with idiopathic myelofibrosis. This disorder, formerly called agnogenic myeloid metaplasia, is a myeloproliferative disease with clonal origin in a malignant pluripotent stem cell. Bone marrow fibrosis is a secondary process. The patient was thought to be heterozygous since her only son had normal G6PD. That she showed severe G6PD deficiency was taken to indicate that the normal X chromosome was active in the original cell that underwent malignancy.

G6PD THESSALY. See Stamatoyannopoulos et al. (1970).

G6PD TITTERI. See Benabadji et al. (1978).

G6PD TITUSVILLE. Csepreghy et al. (1989) described a new G6PD variant in a 7-month-old black male and his mother. The proband had had a transient hemolytic episode.

G6PD TOKUSHIMA. See Miwa et al. (1976).

G6PD TOKYO. See Miwa et al. (1976).

G6PD TORONTO. See Crookston et al. (1973).

G6PD TORRANCE. See Tanaka and Beutler (1969).

G6PD TOULOUSE. See Vergnes et al. (1974).

G6PD 'TRAPANI'. See Sansone et al. (1981) and Perroni et al. (1982).

G6PD TRINACRIA. See Sansone et al. (1977).

G6PD TRIPLER. See Engstrom and Beutler (1970).

G6PD TSUKUI. See Ogura et al. (1988).

G6PD TUBINGEN. See Benohr and Waller (1970).

G6PD TURSI. See Viglietto et al. (1990) and Calabro et al. (1990).

G6PD UBE. See Nakashima et al. (1977).

G6PD UNION. See Yoshida et al. (1970).

G6PD 'UNION-MARKHAM'. See Stamatoyannopoulos et al. (1971).

G6PD 'UNNAMED'. See Othieno-Obel (1972).

G6PD 'VARADERO'. See Estrada et al. (1982).

G6PD VELLETRI. See Mandelli et al. (1977).

G6PD VIENTIANE. See Kahn et al. (1978).

G6PD 'VIN FU'. See Toncheva (1986).

G6PD WAKAYAMA. This variant was found in a 16-month-old boy with 4.5% of normal enzyme activity and mild hemolytic anemia (Miwa et al., 1978). Electrophoretically, it is slow-moving like G6PD Kurume, from which it differs by a normal pH curve. In addition to the 4 slow variants reported by Miwa et al. (1978), 5 had previously been reported: Alhambra, Atlanta, Hong Kong Pokfulam, Manchester, and Tokyo. G6PD WASHINGTON. McCurdy, P. R.: unpublished, 1975.

G6PD WATERLOO. Beutler, E. and Phyliky, R. L.: unpublished, 1978.

G6PD WAYNE. See Ravindranath and Beutler (1987).

G6PD WEST BENGAL. See Azevedo et al. (1968).

G6PD WEST TOWN. This variant causes chronic nonspherocytic anemia which is compensated except following infections or exposure to an oxidant drug (Honig et al., 1979).

G6PD WESTERN. Yoshida, A. and Baur, E.: unpublished, 1975.

G6PD WEWAK. See Chockkalingam et al. (1982).

G6PD WORCESTER. Snyder et al. (1970) described a family in which a new variant form of G6PD was associated with congenital nonspherocytic hemolytic anemia and optic atrophy in 3 males related as first cousins once removed. Blindness developed rapidly in the teens.

G6PD 'WROCLAW'. See Kwiatkowska and Kacprzak-Bergman (1971).

G6PD YAMAGUCHI. This variant was found in an 8-year-old boy who had 3.5% of normal enzyme activity and moderate hemolytic anemia (Miwa et al., 1978). Electrophoretically, it is slow-moving, like G6PD Kurume, from which it differs by high Km NADP, high deamino-NADP utilization, and an abnormal pH curve of a different type (with narrow peak at pH 8.76).

G6PD YANGORU. See Chockkalingam et al. (1982).

G6PD YOKOHAMA. See Miwa et al. (1978).

G6PD 'ZAEHRINGEN'. See Witt and Yoshioka (1972).

G6PD ZAKATALY. See Krasnopolskaya et al. (1977).

G6PD ZHITOMIR. See Shatskaya et al. (1976).


See Also:

Balinsky et al. (1973); Benohr et al. (1971); Beutler et al. (1991); Beutler et al. (1991); Beutler (1975); Boyer et al. (1962); Cazzola and Bergamaschi (1998); Chan and Todd (1972); Chang et al. (1992); Chockkalingam et al. (1982); Cooper et al. (1975); Corash et al. (1980); Epstein (1969); Fiorelli et al. (1989); Fite et al. (1983); Francke et al. (1974); Friedman and Trager (1981); Gahr et al. (1976); Gourdin et al. (1972); Hirono and Beutler (1989); Johnston et al. (1975); Kahn et al. (1975); Kahn et al. (1975); Kirkman et al. (1964); Kirkman et al. (1964); Luzzatto (1974); Martin et al. (1979); McCurdy and Mahmood (1970); McCurdy (1971); Meloni et al. (1990); Miwa et al. (1978); Modiano et al. (1979); Morisaki et al. (1983); O'Brien (1980); Panich et al. (1972); Panich (1974); Persico et al. (1981); Persico et al. (1986); Porter et al. (1962); Roth et al. (1983); Shatskaya et al. (1976); Shatskaya et al. (1980); Shows and Brown (1975); Stamatoyannopoulos et al. (1970); Takizawa et al. (1987); Toniolo et al. (1988); Yoshida (1967); Yoshida (1967)

REFERENCES

  1. Adam, A., Tippett, P., Gavin, J., Noades, J., Sanger, R., Race, R. R. The linkage relation of Xg to G6PD in Israelis: the evidence of a second series of families. Ann. Hum. Genet. 30: 211-218, 1967. [PubMed: 6032005] [Full Text: https://doi.org/10.1111/j.1469-1809.1967.tb00022.x]

  2. Ahluwalia, A., Corcoran, C. M., Vulliamy, T. J., Ishwad, C. S., Naidu, J. M., Argusti, A., Stevens, D. J., Mason, P. J., Luzzatto, L. G6PD Kalyan and G6PD Kerala; two deficient variants in India caused by the same 317 glu-to-lys mutation. Hum. Molec. Genet. 1: 209-210, 1992. [PubMed: 1303182] [Full Text: https://doi.org/10.1093/hmg/1.3.209]

  3. Aksoy, K., Yuregir, G. T., Dikmen, N., Unlukurt, I. Three new G6PD variants, G6PD Adana, G6PD Samandag, and G6PD Balcali in Cukurova, Turkey. Hum. Genet. 76: 199-201, 1987. [PubMed: 3610149] [Full Text: https://doi.org/10.1007/BF00284922]

  4. Alfinito, F., Cimmino, A., Ferraro, F., Cubellis, M. V., Vitagliano, L., Francese, M., Zagari, A., Rotoli, B., Filosa, S., Martini, G. Molecular characterization of G6PD deficiency in Southern Italy: heterogeneity, correlation genotype-phenotype and description of a new variant (G6PD Neapolis). Brit. J. Haemat. 98: 41-46, 1997. [PubMed: 9233561] [Full Text: https://doi.org/10.1046/j.1365-2141.1997.1512967.x]

  5. Alperin, J. B., Mills, G. C. New variants of glucose-6-phosphate dehydrogenase (G6PD). (Abstract) Clin. Res. 20: 76, 1972.

  6. Azevedo, E., Kirkman, H. N., Morrow, A. C., Motulsky, A. G. Variants of red cell glucose-6-phosphate dehydrogenase among Asiatic Indians. Ann. Hum. Genet. 31: 373-379, 1968. [PubMed: 5673160] [Full Text: https://doi.org/10.1111/j.1469-1809.1968.tb00569.x]

  7. Azevedo, E. S., Yoshida, A. Brazilian variant of glucose-6-phosphate dehydrogenase (GD Minas Gerais). Nature 222: 380-382, 1969. [PubMed: 5782116] [Full Text: https://doi.org/10.1038/222380a0]

  8. Babalola, A. O. G., Beetlestone, J. G., Luzzatto, L. Genetic variants of human erythrocyte glucose-6-phosphate dehydrogenase: kinetic and thermodynamic parameters of variants A, B, and A- in relation to quaternary structure. J. Biol. Chem. 251: 2993-3002, 1976. [PubMed: 5448]

  9. Balinsky, D., Cayanis, E., Carter, G., Jenkins, T., Bersohn, I. A new variant of human erythrocyte glucose-6-phosphate dehydrogenase: G6PD Port Elizabeth. Int. J. Biochem. 4: 235-244, 1973.

  10. Balinsky, D., Gomperts, E., Cayanis, E., Jenkins, T., Bryer, D., Bersohn, I., Metz, J. Glucose 6-phosphate dehydrogenase Johannesburg: a new variant with reduced activity in a patient with congenital non-spherocytic haemolytic anaemia. Brit. J. Haemat. 25: 385-392, 1973. [PubMed: 4147554] [Full Text: https://doi.org/10.1111/j.1365-2141.1973.tb01749.x]

  11. Balinsky, D., Rootman, A. J., Nurse, G. T., Cayanis, E., Lane, A., Jenkins, T., Bersohn, I. G6PD Kuanyama: a new variant of human erythrocyte glucose 6-phosphate dehydrogenase showing slower than normal electrophoretic mobility. S. Afr. J. Med. Sci. 39: 5-13, 1974. [PubMed: 4153368]

  12. Barisic, M., Korac, J., Pavlinac, I., Krzelj, V., Marusic, E., Vulliamy, T., Terzic, J. Characterization of G6PD deficiency in southern Croatia: description of a new variant, G6PD Split. J. Hum. Genet. 50: 547-549, 2005. [PubMed: 16143877] [Full Text: https://doi.org/10.1007/s10038-005-0292-2]

  13. Barretto, O. C., Nonoyama, K. Gd(+) Cuiaba, a new rare glucose-6-phosphate dehydrogenase variant presenting normal activity. Hum. Genet. 77: 201-202, 1987. [PubMed: 3653894] [Full Text: https://doi.org/10.1007/BF00272394]

  14. Batischev, A. I., Chernyak, N. B., Torakev, Y. N. Detection of a new abnormal variant of glucose-6-phosphate dehydrogenase in human red cells. Biull. Eksp. Biol. Med. 84: 728-731, 1977. [PubMed: 23188]

  15. Ben-Bassat, J., Ben-Ishay, D. Hereditary hemolytic anemia associated with glucose-6-phosphate dehydrogenase deficiency (Mediterranean type). Isr. J. Med. Sci. 5: 1053-1059, 1969. [PubMed: 5369703]

  16. Benabadji, M., Merad, F., Benmoussa, M., Trabuchet, G., Junien, C., Dreyfus, J. C., Kaplan, J. C. Heterogeneity of glucose-6-phosphate dehydrogenase deficiency in Algeria. Hum. Genet. 40: 177-184, 1978. [PubMed: 624546] [Full Text: https://doi.org/10.1007/BF00272298]

  17. Benohr, H. C., Klumpp, F., Waller, H. D. Glucose-6-phosphat-Dehydrogenase Typ Schwaben. Dtsch. Med. Wschr. 96: 1029-1032, 1971. [PubMed: 5580403] [Full Text: https://doi.org/10.1055/s-0028-1108377]

  18. Benohr, H. C., Waller, H. D., Arnold, H., Blume, K. G., Lohr, G. W. Glucose-6-P-Dehydrogenase Typ Bodensee (eine neue Enzymvariante). Klin. Wschr. 49: 1058-1062, 1971. [PubMed: 5115324] [Full Text: https://doi.org/10.1007/BF01732914]

  19. Benohr, H. C., Waller, H. D. Eigenschaften der Glucose-6-p-dehydrogenase, Typ Tubingen. Klin. Wschr. 48: 71-74, 1970. [PubMed: 4398790] [Full Text: https://doi.org/10.1007/BF01484619]

  20. Beutler, E., Grooms, A. M., Morgan, S. K., Trinidad, F. Chronic severe hemolytic anemia due to G-6-PD Charleston: a new deficiency variant. J. Pediat. 80: 1005-1009, 1972. [PubMed: 4402065] [Full Text: https://doi.org/10.1016/s0022-3476(72)80014-3]

  21. Beutler, E., Hartman, K., Gelbart, T., Forman, L. G-6-PD Walter Reed: possible insight into 'structural' NADP in G-6-PD. Am. J. Hemat. 23: 25-30, 1986. [PubMed: 3740052] [Full Text: https://doi.org/10.1002/ajh.2830230105]

  22. Beutler, E., Keller, J. W., Matsumoto, F. A new glucose 6-phosphate dehydrogenase (G6PD) variant associated with nonspherocytic hemolytic anemia: G6PD Atlanta. I.R.C.S. 4: 479, 1976.

  23. Beutler, E., Kuhl, W., Gelbart, T., Forman, L. DNA sequence abnormalities of human glucose-6-phosphate dehydrogenase variants. J. Biol. Chem. 266: 4145-4150, 1991. [PubMed: 1999409]

  24. Beutler, E., Kuhl, W., Ramirez, E., Lisker, R. Some Mexican glucose-6-phosphate dehydrogenase variants revisited. Hum. Genet. 86: 371-374, 1991. [PubMed: 1999339] [Full Text: https://doi.org/10.1007/BF00201837]

  25. Beutler, E., Kuhl, W., Saenz, G. F., Rodriguez, W. Mutation analysis of glucose-6-phosphate dehydrogenase (G6PD) variants in Costa Rica. Hum. Genet. 87: 462-464, 1991. [PubMed: 1879833] [Full Text: https://doi.org/10.1007/BF00197169]

  26. Beutler, E., Kuhl, W., Vives-Corrons, J.-L., Prchal, J. T. Molecular heterogeneity of glucose-6-phosphate dehydrogenase A-. Blood 74: 2550-2555, 1989. [PubMed: 2572288]

  27. Beutler, E., Kuhl, W. Linkage between a PvuII restriction fragment length polymorphism and G6PD A- (202A/376G): evidence for a single origin of the common G6PD A- mutation. Hum. Genet. 85: 9-11, 1990. [PubMed: 1972698] [Full Text: https://doi.org/10.1007/BF00276317]

  28. Beutler, E., Kuhl, W. The NT 1311 polymorphism of G6PD: G6PD Mediterranean mutation may have originated independently in Europe and Asia. Am. J. Hum. Genet. 47: 1008-1012, 1990. [PubMed: 1978554]

  29. Beutler, E., Mathai, C. K., Smith, J. E. Biochemical variants of glucose-6-phosphate dehydrogenase giving rise to congenital nonspherocytic hemolytic disease. Blood 31: 131-150, 1968. [PubMed: 5643703]

  30. Beutler, E., Matsumoto, F., Daiber, A. Nonspherocytic hemolytic anemia due to G-6-PD Panama. I.R.C.S. 2: 1389, 1974.

  31. Beutler, E., Matsumoto, F. A new glucose 6-phosphate dehydrogenase variant: G6PD (-) Los Angeles. I.R.C.S. 5: 89, 1977.

  32. Beutler, E., Rosen, R. Nonspherocytic congenital hemolytic anemia due to a new G-6-PD variant: G-6-PD Alhambra. Pediatrics 45: 230-235, 1970. [PubMed: 5413384]

  33. Beutler, E., Westwood, B., Prchal, J. T., Vaca, G., Bartsocas, C. S., Baronciani, L. New glucose-6-phosphate dehydrogenase mutations from various ethnic groups. Blood 80: 255-256, 1992. [PubMed: 1611091]

  34. Beutler, E., Yoshida, A. Human glucose-6-phosphate dehydrogenase variants: a supplementary tabulation. Ann. Hum. Genet. 37: 151-156, 1973. [PubMed: 4768554] [Full Text: https://doi.org/10.1111/j.1469-1809.1973.tb01823.x]

  35. Beutler, E. Glucose 6-phosphate dehydrogenase deficiency, a new Indian variant, G6PD Jammu. In: Sen, N. N.; Basu, A. K. (eds.): Trends in Haematology. Calcutta: Chatterjea Memorial Committee (pub.) 1975. Pp. 279-283.

  36. Beutler, E. Selectivity of proteases as a basis for tissue distribution of enzymes in hereditary deficiencies. Proc. Nat. Acad. Sci. 80: 3767-3768, 1983. [PubMed: 6344088] [Full Text: https://doi.org/10.1073/pnas.80.12.3767]

  37. Beutler, E. Glucose-6-phosphate dehydrogenase deficiency. New Eng. J. Med. 324: 169-174, 1991. [PubMed: 1984194] [Full Text: https://doi.org/10.1056/NEJM199101173240306]

  38. Boivin, P., Galand, C. Nouvelles variantes de la glucose-6-phosphate dehydrogenase erythrocytaire. Rev. Franc. Etud. Clin. Biol. 13: 30-39, 1968. [PubMed: 5653554]

  39. Botha, M. C., Dern, R. J., Mitchell, M., West, C., Beutler, E. G6PD Capetown, a variant of glucose-6-phosphate dehydrogenase. Am. J. Hum. Genet. 21: 547-551, 1969. [PubMed: 5365756]

  40. Boyer, S. H., Graham, J. B. Linkage between the X chromosome loci for glucose-6-phosphate dehydrogenase electrophoretic variation and hemophilia A. Am. J. Hum. Genet. 17: 320-324, 1965. [PubMed: 17948498]

  41. Boyer, S. H., Porter, I. H., Weilbaecher, R. G. Electrophoretic heterogeneity of glucose-6-phosphate dehydrogenase and its relationship to enzyme deficiency in man. Proc. Nat. Acad. Sci. 48: 1868-1876, 1962. [PubMed: 14014720] [Full Text: https://doi.org/10.1073/pnas.48.10.1868]

  42. Busch, D., Boie, K. Glucose-6-phosphate-dehydrogenase-Defect in Deutschland. II. Bigenschabten des Enzyms (Typ Freiburg). Klin. Wschr. 48: 74-78, 1970. [PubMed: 4398791] [Full Text: https://doi.org/10.1007/BF01484620]

  43. Calabro, V., Giacobbe, A., Vallone, D., Montanaro, V., Cascone, A., Filosa, S., Battistuzzi, G. Genetic heterogeneity at the glucose-6-phosphate dehydrogenase locus in southern Italy: a study on a population from the Matera district. Hum. Genet. 86: 49-53, 1990. [PubMed: 2253938] [Full Text: https://doi.org/10.1007/BF00205171]

  44. Calabro, V., Mason, P. J., Filosa, S., Civitelli, D., Cittadella, R., Tagarelli, A., Martini, G., Brancati, C., Luzzatto, L. Genetic heterogeneity of glucose-6-phosphate dehydrogenase deficiency revealed by single-strand conformation and sequence analysis. Am. J. Hum. Genet. 52: 527-536, 1993. [PubMed: 8447319]

  45. Cappadoro, M., Giribaldi, G., O'Brien, E., Turrini, F., Mannu, F., Ulliers, D., Simula, G., Luzzatto, L., Arese, P. Early phagocytosis of glucose-6-phosphate dehydrogenase (G6PD)-deficient erythrocytes parasitized by Plasmodium falciparum may explain malaria protection in G6PD deficiency. Blood 92: 2527-2534, 1998. [PubMed: 9746794]

  46. Cappellini, M. D., Fiorelli, G. Glucose-6-phosphate dehydrogenase deficiency. Lancet 371: 64-74, 2008. [PubMed: 18177777] [Full Text: https://doi.org/10.1016/S0140-6736(08)60073-2]

  47. Cappellini, M. D., Sampietro, M., Toniolo, D., Carandina, G., Pittalis, S., Martinez di Montemuros, F., Tavazzi, D., Fiorelli, G. Biochemical and molecular characterization of a new sporadic glucose-6-phosphate dehydrogenase variant described in Italy: G6PD Modena. Brit. J. Haemat. 87: 209-211, 1994. [PubMed: 7947250] [Full Text: https://doi.org/10.1111/j.1365-2141.1994.tb04896.x]

  48. Carandina, G., Moretto, E., Zecchi, G., Conighi, C. Glucose 6-phosphate dehydrogenase Ferrara. A new variant of G6PD identified in Northern Italy. Acta Haemat. 56: 116-122, 1976. [PubMed: 823756] [Full Text: https://doi.org/10.1159/000207927]

  49. Castro, A. M., Snyder, L. M. G6PD San Jose: a new variant characterized by NADPH inhibition studies. Humangenetik 21: 361-363, 1974. [PubMed: 4151903] [Full Text: https://doi.org/10.1007/BF00273375]

  50. Cayanis, E., Gomperts, E. D., Balinsky, D., Disler, P., Myers, A. G6PD Hillbrow: a new variant of glucose-6-phosphate dehydrogenase associated with drug-induced haemolytic anaemia. Brit. J. Haemat. 30: 343-350, 1975. [PubMed: 1201217] [Full Text: https://doi.org/10.1111/j.1365-2141.1975.tb00550.x]

  51. Cayanis, E., Lane, A. B., Jenkins, T., Nurse, G. T., Balinsky, D. Glucose-6-phosphate dehydrogenase Porbandar: a new slow variant with slightly reduced activity in a South African family of Indian descent. Biochem. Genet. 15: 765-773, 1977. [PubMed: 921743] [Full Text: https://doi.org/10.1007/BF00484102]

  52. Cazzola, M., Bergamaschi, G. X-linked Wiskott-Aldrich syndrome in a girl. (Letter) New Eng. J. Med. 338: 1850 only, 1998. [PubMed: 9634367] [Full Text: https://doi.org/10.1056/NEJM199806183382515]

  53. Cederbaum, A. I., Beutler, E. Nonspherocytic hemolytic anemia due to G6PD Grand Prairie. I.R.C.S. 3: 579, 1975.

  54. Chalvam, R., Kedar, P. S., Colah, R. B., Ghosh, K., Mukherjee, M. B. A novel R198H mutation in the glucose-6-phosphate dehydrogenase gene in the tribal groups of the Nilgiris in southern India. J. Hum. Genet. 53: 181-184, 2008. [PubMed: 18043863] [Full Text: https://doi.org/10.1007/s10038-007-0225-3]

  55. Chalvam, R., Mukherjee, M. B., Colah, R. B., Mohanty, D., Ghosh, K. G6PD Namoru (208T-C) is the major polymorphic variant in the tribal populations in southern India. (Letter) Brit. J. Haemat. 136: 512-513, 2007. [PubMed: 17233850] [Full Text: https://doi.org/10.1111/j.1365-2141.2006.06463.x]

  56. Chan, T. K., Todd, D., Lai, M. C. S. Glucose 6-phosphate dehydrogenase: identity of erythrocyte and leukocyte enzyme with report of a new variant in Chinese. Biochem. Genet. 6: 119-124, 1972. [PubMed: 4666752] [Full Text: https://doi.org/10.1007/BF00486396]

  57. Chan, T. K., Todd, D. Characteristics and distribution of glucose-6-phosphate dehydrogenase-deficient variants in South China. Am. J. Hum. Genet. 24: 475-484, 1972. [PubMed: 5031985]

  58. Chang, J.-G., Chiou, S.-S., Perng, L.-I., Chen, T.-C., Liu, T.-C., Lee, L.-S., Chen, P.-H., Tang, T. K. Molecular characterization of glucose-6-phosphate dehydrogenase (G6PD) deficiency by natural and amplification created restriction sites: five mutations account for most G6PD deficiency cases in Taiwan. Blood 80: 1079-1082, 1992. [PubMed: 1323345]

  59. Chen, E. Y., Cheng, A., Lee, A., Kuang, W.-J., Hillier, L., Green, P., Schlessinger, D., Ciccodicola, A., D'Urso, M. Sequence of human glucose-6-phosphate dehydrogenase cloned in plasmids and a yeast artificial chromosome. Genomics 10: 792-800, 1991. [PubMed: 1889820] [Full Text: https://doi.org/10.1016/0888-7543(91)90465-q]

  60. Chen, H.-L., Huang, M.-J., Huang, C.-S., Tang, T. K. G6PD NanKang (517 T-to-C; 173 phe-to-leu): a new Chinese G6PD variant associated with neonatal jaundice. Hum. Hered. 46: 201-204, 1996. [PubMed: 8807322] [Full Text: https://doi.org/10.1159/000154354]

  61. Cherniak, N. B., Batischev, A. I., Lanzina, N. V., Tokarev, Y. N., Alexeyev, G. A. Electrophoretic and kinetic properties of glucose-6-phosphate dehydrogenase from erythrocytes or patients with hemolytic anemia, related to deficiency of the enzyme activity. Vopr. Med. Khim. 23: 166-171, 1977.

  62. Childs, B., Zinkham, W., Browne, E. A., Kimbro, E. L., Torbert, J. V. A genetic study of a defect in glutathione metabolism of the erythrocytes. Bull. Johns Hopkins Hosp. 102: 21-37, 1958. [PubMed: 13500095]

  63. Chiu, D. T. Y., Zuo, L., Chao, L., Chen, E., Louie, E., Lubin, B., Liu, T. Z., Du, C. S. Molecular characterization of glucose-6-phosphate dehydrogenase (G6PD) deficiency in patients of Chinese descent and identification of new base substitutions in the human G6PD gene. Blood 81: 2150-2154, 1993. [PubMed: 8471773]

  64. Chockkalingam, K., Board, P. G., Breguet, G. Glucose-6-phosphate dehydrogenase variants of Bali Island (Indonesia). Hum. Genet. 60: 60-62, 1982. [PubMed: 7076249] [Full Text: https://doi.org/10.1007/BF00281265]

  65. Chockkalingam, K., Board, P. G., Nurse, G. T. Glucose-6-phosphate dehydrogenase deficiency in Papua New Guinea: the description of 13 new variants. Hum. Genet. 60: 189-192, 1982. [PubMed: 7076260] [Full Text: https://doi.org/10.1007/BF00569710]

  66. Chockkalingam, K., Board, P. G. Further evidence for heterogeneity of glucose-6-phosphate dehydrogenase deficiency in Papua New Guinea. Hum. Genet. 56: 209-212, 1980. [PubMed: 7450778] [Full Text: https://doi.org/10.1007/BF00295696]

  67. Cianfriglia, M., Miggiano, V. C., Meo, T., Muller, H. J., Muller, E., Battistuzzi, G. Evidence for synteny between the rabbit gene loci coding for HPRT, PGK and G6PD in mouse-rabbit somatic cell hybrids. (Abstract) Cytogenet. Cell Genet. 25: 142, 1979.

  68. Cohn, J., Carter, N., Warburg, M. Glucose-6-phosphate dehydrogenase deficiency in a native Danish family: a new variant. Scand. J. Haemat. 23: 403-406, 1979. [PubMed: 542803] [Full Text: https://doi.org/10.1111/j.1600-0609.1979.tb02740.x]

  69. Colonna-Romano, S., Iolascon, A., Lippo, S., Pinto, L., Cutillo, S., Battistuzzi, G. Genetic heterogeneity at the glucose-6-phosphate dehydrogenase locus in southern Italy: a study on the population of Naples. Hum. Genet. 69: 228-232, 1985. [PubMed: 3980015] [Full Text: https://doi.org/10.1007/BF00293030]

  70. Cooper, D. W., Johnston, P. G., Murtagh, C. E., Sharman, G. B., Vandeberg, J. L., Poole, W. E. Sex-linked isozymes and sex-chromosome evolution and inactivation in kangaroos. In: Markert, C. L. (ed.): Isozymes. Vol. 3. Developmental Biology. New York: Academic Press (pub.) 1975. Pp. 559-573.

  71. Corash, L., Spielberg, S., Bartsocas, C., Boxer, L., Steinherz, R., Sheetz, M., Egan, M., Schlessleman, J., Schulman, J. D. Reduced chronic hemolysis during high-dose vitamin E administration in Mediterranean-type glucose-6-phosphate dehydrogenase deficiency. New Eng. J. Med. 303: 416-420, 1980. [PubMed: 7393270] [Full Text: https://doi.org/10.1056/NEJM198008213030802]

  72. Corcoran, C. M., Calabro, V., Tamagnini, G., Town, M., Haidar, B., Vulliamy, T. J., Mason, P. J., Luzzatto, L. Molecular heterogeneity underlying the G6PD Mediterranean phenotype. Hum. Genet. 88: 688-690, 1992. [PubMed: 1551674] [Full Text: https://doi.org/10.1007/BF02265298]

  73. Costa, E., Cabeda, J. M., Vieira, E., Pinto, R., Pereira, S. A., Ferraz, L., Santos, R., Barbot, J. Glucose-6-phosphate dehydrogenase Aveiro: a de novo mutation associated with chronic nonspherocytic hemolytic anemia. Blood 95: 1499-1501, 2000. [PubMed: 10666231]

  74. Crookston, J. H., Yoshida, A., Lin, M., Booser, D. J. G6PD Toronto. Biochem. Genet. 8: 259-265, 1973. [PubMed: 4701992] [Full Text: https://doi.org/10.1007/BF00486178]

  75. Csepreghy, M., Hall, M. K., Berkow, R. L., Jackson, S., Prchal, J. T. Characterization of a new G6PD variant: G6PD Titusville. Am. J. Med. Sci. 297: 114-117, 1989. [PubMed: 2919631] [Full Text: https://doi.org/10.1097/00000441-198902000-00007]

  76. Csepreghy, M., Yeilding, A., Lilly, M., Hall, K., Scott, C. W., Prchal, J. T. Characterization of a new glucose-6-phosphate dehydrogenase variant: G6PD Central City. Am. J. Hemat. 28: 61-62, 1988. [PubMed: 3369438] [Full Text: https://doi.org/10.1002/ajh.2830280114]

  77. D'Urso, M., Luzzatto, L., Perroni, L., Ciccodicola, A., Gentile, G., Peluso, I., Persico, M. G., Pizzella, T., Toniolo, D., Vulliamy, T. J. An extensive search for RFLP in the human glucose-6-phosphate dehydrogenase locus has revealed a silent mutation in the coding sequence. Am. J. Hum. Genet. 42: 735-741, 1988. [PubMed: 2895981]

  78. Davidson, R. G., Nitowsky, H. M., Childs, B. Demonstration of two populations of cells in the human female heterozygous for glucose-6-phosphate dehydrogenase variants. Proc. Nat. Acad. Sci. 50: 481-485, 1963. [PubMed: 14067093] [Full Text: https://doi.org/10.1073/pnas.50.3.481]

  79. De Flora, A., Morelli, A., Benatti, U., Giuntini, P., Ferraris, A. M., Galiano, S., Ravazzolo, R., Gaetani, G. F. G6PD Napoli and Ferrara II: two new glucose-6-phosphate dehydrogenase variants having similar characteristics but different intracellular lability and specific activity. Brit. J. Haemat. 48: 417-423, 1981. [PubMed: 7259990] [Full Text: https://doi.org/10.1111/j.1365-2141.1981.tb02733.x]

  80. De Vita, G., Alcalay, M., Sampietro, M., Cappelini, M. D., Fiorelli, G., Toniolo, D. Two point mutations are responsible for G6PD polymorphism in Sardinia. Am. J. Hum. Genet. 44: 233-240, 1989. [PubMed: 2912069]

  81. Dern, R. J., McCurdy, P. R., Yoshida, A. A new structural variant of glucose-6-phosphate dehydrogenase with a high production rate (G6PD Hektoen). J. Lab. Clin. Med. 73: 283-290, 1969. [PubMed: 4974311]

  82. Du, C. S., Xu, Y. K., Hua, X. Y., Wu, Q. L., Liu, L. B. Glucose-6-phosphate dehydrogenase variants and their frequency in Guangdong, China. Hum. Genet. 80: 385-388, 1988. [PubMed: 3198117] [Full Text: https://doi.org/10.1007/BF00273657]

  83. Du, C. S. Studies on erythrocyte glucose-6-phosphate dehydrogenase variants in Chinese. I. Gd(-) Lizu-Baisha. Chin. Med. J. 61: 729-732, 1981. [PubMed: 6805883]

  84. Du, C., Xu, Y., Hua, X., Wu, Q., Liu, L., Wu, M. Studies on erythrocyte glucose-6-phosphate dehydrogenase variants in Chinese. III: Gd(-) Miaozu-Baisha. Acta Genet. Sin. 11: 153-158, 1984. [PubMed: 6805883]

  85. Du, C.-S., Hua, X.-Y., Wu, Q.-L., Li, C.-Q., Zheng, J.-F., Li, H.-L. Studies on erythrocyte glucose-6-phosphate dehydrogenase variants in Chinese. IV. Gd(-) Gaohe associated with paroxysmal nocturnal hemoglobinuria. Chinese J. Pathophysiol. 1: 12-15, 1985.

  86. Eber, S. W., Gahr, M., Schroter, W. Glucose-6-phosphate dehydrogenase (G6PD) Iserlohn and G6PD Regensburg: two new severe enzyme defects in German families. Blut 51: 109-115, 1985. [PubMed: 3849316] [Full Text: https://doi.org/10.1007/BF00320119]

  87. Echard, G., Gillois, M. G6PD--alpha-GAL-PGK--HPRT synteny in the rabbit, Oryctolagus cunniculus. (Abstract) Cytogenet. Cell Genet. 25: 148-149, 1979.

  88. Efferth, T., Bachli, E. B., Schwarzl, S. M., Goede, J. S., West, C., Smith, J. C., Beutler, E. Glucose-6-phosphate dehydrogenase (G6PD) deficiency-type Zurich: a splice site mutation as an uncommon mechanism producing enzyme deficiency. (Letter) Blood 104: 2608 only, 2004. [PubMed: 15466166] [Full Text: https://doi.org/10.1182/blood-2004-06-2135]

  89. Elizondo, J., Saenz, G. F., Paez, C. A., Ramon, M., Garcia, M., Gutierrez, A., Estrada, M. G6PD Puerto Limon: a new deficient variant of glucose-6-phosphate dehydrogenase associated with congenital nonspherocytic hemolytic anemia. Hum. Genet. 62: 110-112, 1982. [PubMed: 7160841] [Full Text: https://doi.org/10.1007/BF00282295]

  90. Engstrom, P. F., Beutler, E. G-6-PD Tripler: a unique variant associated with chronic hemolytic disease. Blood 36: 10-13, 1970. [PubMed: 5421739]

  91. Epstein, C. J. Mammalian oocytes: X-chromosome activity. Science 163: 1078-1079, 1969. [PubMed: 5764873] [Full Text: https://doi.org/10.1126/science.163.3871.1078]

  92. Ermakov, N. V., Chernyak, N. B., Tokarev, Y. N. Properties of new variant of glucose-6-phosphate dehydrogenase (Regar variant): glucose metabolism in erythrocytes containing abnormal enzyme. Biokhimiia 48: 577-583, 1983.

  93. Estrada, M., Garcia, M., Gutierrez, A., Quintero, I., Gonzalez, R. G6PD Varadero. Vox Sang. 43: 102-104, 1982. [PubMed: 7123903] [Full Text: https://doi.org/10.1111/j.1423-0410.1982.tb00533.x]

  94. Fairbanks, V. F., Nepo, A. G., Beutler, E., Dickson, E. R., Honig, G. Glucose-6-phosphate dehydrogenase variants: reexamination of G6PD Chicago and Cornell and a new variant (G6PD Pea Ridge) resembling G6PD Chicago. Blood 55: 216-220, 1980. [PubMed: 7353072]

  95. Faust, C. J., Levinson, B., Gitschier, J., Herman, G. E. Extension of the physical map in the region of the mouse X chromosome homologous to human Xq28 and identification of an exception to conserved linkage. Genomics 13: 1289-1295, 1992. [PubMed: 1354645] [Full Text: https://doi.org/10.1016/0888-7543(92)90048-w]

  96. Feldman, R., Gromisch, D. S., Luhby, A. L., Beutler, E. Congenital nonspherocytic hemolytic anemia due to glucose-6-phosphate dehydrogenase East Harlem: a new deficient variant. J. Pediat. 90: 89-91, 1977. [PubMed: 830903] [Full Text: https://doi.org/10.1016/s0022-3476(77)80774-9]

  97. Fernandez, M., Fairbanks, V. F. Glucose-6-phosphate dehydrogenase deficiency in the Philippines: report of a new variant--G6PD Panay. Mayo Clin. Proc. 43: 645-660, 1968. [PubMed: 5710590]

  98. Filosa, E. Personal Communication. La Jolla, Calif. 1989.

  99. Filosa, S., Calabro, V., Vallone, D., Poggi, V., Mason, P., Pagnini, D., Alfinito, F., Rotoli, B., Martini, G., Luzzatto, L., Battistuzzi, G. Molecular basis of chronic non-spherocytic haemolytic anaemia: a new G6PD variant (393arg-to-his) with abnormal K(m) G6P and marked in vivo instability. Brit. J. Haemat. 80: 111-116, 1992. [PubMed: 1536798] [Full Text: https://doi.org/10.1111/j.1365-2141.1992.tb06409.x]

  100. Filosa, S., Giacometti, N., Wangwei, C., De Mattia, D., Pagnini, D., Alfinito, F., Schettini, F., Luzzatto, L., Martini, G. Somatic-cell selection is a major determinant of the blood-cell phenotype in heterozygotes for glucose-6-phosphate dehydrogenase mutations causing severe enzyme deficiency. Am. J. Hum. Genet. 59: 887-895, 1996. [PubMed: 8808605]

  101. Fiorelli, G., Manoussakis, C., Sampietro, M., Pittalis, S., Guglielmino, C. R., Cappellini, M. D. Different polymorphic variants of glucose-6-phosphate dehydrogenase (G6PD) in Italy. Ann. Hum. Genet. 53: 229-236, 1989. [PubMed: 2596828] [Full Text: https://doi.org/10.1111/j.1469-1809.1989.tb01789.x]

  102. Fite, E., Morell, F., Zuazu, J., Julia, A., Morera, J. Leucocyte glucose-6-phosphate dehydrogenase deficiency and necrotizing pneumonia. Europ. J. Resp. Dis. 64: 150-154, 1983. [PubMed: 6832252]

  103. Francke, U., Bakay, B., Connor, J. D., Coldwell, J. G., Nyhan, W. L. Linkage relationships of X-linked enzymes glucose-6-phosphate dehydrogenase and hypoxanthine guanine phosphoribosyltransferase. Am. J. Hum. Genet. 26: 512-522, 1974. [PubMed: 4842775]

  104. Friedman, M. J., Trager, W. The biochemistry of resistance to malaria. Sci. Am. 244(3): 154-164, 1981. [PubMed: 6163210] [Full Text: https://doi.org/10.1038/scientificamerican0381-154]

  105. Frigerio, R., Sole, G., Olla, N., Lovicu, M., Passiu, G., Carcassi, U. Cagliari II: a new G-6-PD variant. Haematologica 72: 241-243, 1987. [PubMed: 3114091]

  106. Fujii, H., Miwa, S., Takegawa, S., Takahashi, K., Hirono, A., Takizawa, T., Morisaki, T., Kanno, H., Taguchi, T., Okamura, J. Gd(-) Gifu and Gd(-) Fukuoka: two new variants of glucose-6-phosphate dehydrogenase found in Japan. Hum. Genet. 66: 276-278, 1984. [PubMed: 6714986] [Full Text: https://doi.org/10.1007/BF00286616]

  107. Fujii, H., Miwa, S., Tani, K., Takegawa, S., Fujinami, N., Takahashi, K., Nakayama, S., Konno, M., Sato, T. Glucose 6-phosphate dehydrogenase variants: a unique variant (G6PD Kobe) showed an extremely increased affinity for galactose 6-phosphate and a new variant (G6PD Sapporo) resembling G6PD Pea Ridge. Hum. Genet. 58: 405-407, 1981. [PubMed: 7327562] [Full Text: https://doi.org/10.1007/BF00282823]

  108. Fusco, F., Paciolla, M., Napolitano, F., Pescatore, A., D'Addario, I., Bal, E., Lioi, M. B., Smahl, A., Miano, M. G., Ursini, M. V. Genomic architecture at the incontinentia pigmenti locus favours de novo pathological alleles through different mechanisms. Hum. Molec. Genet. 21: 1260-1271, 2012. [PubMed: 22121116] [Full Text: https://doi.org/10.1093/hmg/ddr556]

  109. Gaetani, G. F., Galiano, S., Melani, C., Miglino, M., Forni, G. L., Napoli, G., Perrone, L., Ferraris, A. M. A new glucose-6-phosphate dehydrogenase variant with congenital nonspherocytic hemolytic anemia (G6PD Genova): biochemical characterization and mosaicism expression in the heterozygote. Hum. Genet. 84: 337-340, 1990. [PubMed: 2307454] [Full Text: https://doi.org/10.1007/BF00196229]

  110. Gahr, M., Bornhalm, D., Schroter, W. Haemolytic anemia due to glucose 6-phosphate dehydrogenase (G6PD) deficiency: demonstration of two new biochemical variants, G6PD Hamm and G6PD Tarsus. Brit. J. Haemat. 33: 363-370, 1976. [PubMed: 1276080] [Full Text: https://doi.org/10.1111/j.1365-2141.1976.tb03552.x]

  111. Gahr, M., Bornhalm, D., Schroter, W. Biochemische Eigenschaften einer neuen Variante des Glucose-6-phosphadehydrogenase (G6PD) Mangels mit Favismus: G6PD Bielefeld. Klin. Wschr. 55: 379-384, 1977. [PubMed: 859288] [Full Text: https://doi.org/10.1007/BF01488623]

  112. Gahr, M., Schroter, W., Sturzenegger, M., Bornhalm, D., Marti, H. R. Glucose 6-phosphate dehydrogenase (G6PD) deficiency in Switzerland. Helv. Paediat. Acta 31: 159-166, 1976. [PubMed: 955939]

  113. Gahr, M., Schroter, W. Glucose 6-phosphate dehydrogenase (G6PD) Hamburg, a new variant with chronic nonspherocytic hemolytic anemia. Europ. J. Clin. Invest. 4: 187-191, 1974. [PubMed: 4838696] [Full Text: https://doi.org/10.1111/j.1365-2362.1974.tb00390.x]

  114. Geerdink, R. A., Horst, R., Staal, G. E. An Iraqi Jewish family with a new red cell glucose 6-phosphate dehydrogenase variant (Gd-Bagdad) and kernicterus. Isr. J. Med. Sci. 9: 1040-1043, 1973. [PubMed: 4753845]

  115. Gellin, J., Benne, F., Renard, C., Vaiman, M., Hors-Cayla, M. C., Gillois, M. Pig gene mapping: synteny, attempt to assign the histocompatibility complex (SLA). (Abstract) Cytogenet. Cell Genet. 25: 159, 1979.

  116. Gomez-Gallego, F., Garrido-Pertierra, A., Bautista, J. M. Structural defects underlying protein dysfunction in human glucose-6-phosphate dehydrogenase A- deficiency. J. Biol. Chem. 275: 9256-9262, 2000. [PubMed: 10734064] [Full Text: https://doi.org/10.1074/jbc.275.13.9256]

  117. Gonzalez, R., Estrada, M., Garcia, M., Gutierrez, A. G6PD Ciudad de la Habana: a new slow variant with deficiency found in a Cuban family. Hum. Genet. 55: 133-135, 1980. [PubMed: 7450750] [Full Text: https://doi.org/10.1007/BF00329141]

  118. Gonzalez, R., Wade, M., Estrada, M., Svarch, E., Colombo, B. G6PD Pinar del Rio: a new variant discovered in a Cuban family. Biochem. Genet. 15: 909-913, 1977. [PubMed: 588239] [Full Text: https://doi.org/10.1007/BF00483987]

  119. Goss, S. J., Harris, H. Gene transfer by means of cell fusion. I. Statistical mapping of the human X-chromosome by analysis of radiation induced gene segregation. J. Cell Sci. 25: 17-37, 1977. [PubMed: 561093] [Full Text: https://doi.org/10.1242/jcs.25.1.17]

  120. Gourdin, D., Vergnes, H., Bouloux, C., Ruffie, J., Gherardi, M. Polymorphism of erythrocyte G6PD in the baboon. Am. J. Phys. Anthrop. 37: 281-288, 1972. [PubMed: 4628608] [Full Text: https://doi.org/10.1002/ajpa.1330370213]

  121. Gray, G. R., Stamatoyannopoulos, G., Naiman, S. C., Kliman, M. R., Klebanoff, S. J., Austin, T., Yoshida, A., Robinson, G. C. G. Neutrophil dysfunction, chronic granulomatous disease, and non-spherocytic haemolytic anaemia caused by complete deficiency of glucose-6-phosphate dehydrogenase. Lancet 302: 530-534, 1973. Note: Originally Volume II. [PubMed: 4125296] [Full Text: https://doi.org/10.1016/s0140-6736(73)92350-7]

  122. Grossman, A., Ramanathan, K., Justice, P., Gordon, J., Shahidi, N. T., Hsia, D. Y. Y. Congenital nonspherocytic hemolytic anemia associated with erythrocyte G-6-PD deficiency in a Negro family. Pediatrics 37: 624-629, 1966. [PubMed: 5930024]

  123. Gutierrez, A., Garcia, M., Estrada, M., Quintero, I., Gonzalez, R. Glucose-6-phosphate dehydrogenase (G6PD) Guantanamo and G6PD Caujeri: two new glucose-6-phosphate dehydrogenase-deficient variants found in Cuba. Biochem. Genet. 25: 231-238, 1987. [PubMed: 3606560] [Full Text: https://doi.org/10.1007/BF00499316]

  124. Hall, K., Schreeder, M. T., Prchal, J. T. G6PD Huntsville: a new glucose-6-phosphate dehydrogenase associated with chronic hemolytic anemia. Hum. Genet. 79: 90-91, 1988. [PubMed: 3366466] [Full Text: https://doi.org/10.1007/BF00291720]

  125. Harkonen, M., Vuopio, P. Red cell glucose-6-phosphate dehydrogenase deficiency in Finland. Ann. Clin. Res. 6: 187-197, 1974. [PubMed: 4154719]

  126. Harley, J. D., Agar, N. S., Yoshida, A. Glucose 6-phosphate dehydrogenase variant Gd(+) Alexandra associated with neonatal jaundice and Gd(-) Camperdown in a young man with lamellar cataracts. J. Lab. Clin. Med. 91: 295-300, 1978. [PubMed: 23402]

  127. Helge, H., Borner, K. Kongenitale nichtsphaerozytare haemolytische Anaemie, Katarakt und Glucose-6-phosphat-dehydrogenase-mangel. Dtsch. Med. Wschr. 91: 1584-1589, 1966. [PubMed: 4380311] [Full Text: https://doi.org/10.1055/s-0028-1111556]

  128. Hirono, A., Beutler, E. Molecular cloning and nucleotide sequence of cDNA for human glucose-6-phosphate dehydrogenase variant A(-). Proc. Nat. Acad. Sci. 85: 3951-3954, 1988. [PubMed: 2836867] [Full Text: https://doi.org/10.1073/pnas.85.11.3951]

  129. Hirono, A., Beutler, E. Alternative splicing of human glucose-6-phosphate dehydrogenase messenger RNA in different tissues. J. Clin. Invest. 83: 343-346, 1989. [PubMed: 2910917] [Full Text: https://doi.org/10.1172/JCI113881]

  130. Hirono, A., Fujii, H., Shima, M., Miwa, S. G6PD Nara: a new class 1 glucose-6-phosphate dehydrogenase variant with an eight amino acid deletion. Blood 82: 3250-3252, 1993. [PubMed: 8241497]

  131. Hirono, A., Kawate, K., Honda, A., Fujii, H., Miwa, S. A single mutation 202G-A in the human glucose-6-phosphate dehydrogenase gene (G6PD) can cause acute hemolysis by itself. (Letter) Blood 99: 1498 only, 2002. [PubMed: 11852882] [Full Text: https://doi.org/10.1182/blood.v99.4.1498]

  132. Hirono, A., Kuhl, W., Gelbart, T., Forman, L., Fairbanks, V. F., Beutler, E. Identification of the binding domain for NADP(+) of human glucose-6-phosphate dehydrogenase by sequence analysis of mutants. Proc. Nat. Acad. Sci. 86: 10015-10017, 1989. [PubMed: 2602358] [Full Text: https://doi.org/10.1073/pnas.86.24.10015]

  133. Hitzeroth, H. W., Bender, K. Age-dependency of somatic selection in South African Negro G-6-PD heterozygotes. Hum. Genet. 58: 338-343, 1981. [PubMed: 7327555] [Full Text: https://doi.org/10.1007/BF00294934]

  134. Honig, G. R., Habacon, E., Vida, L. N., Matsumoto, F., Beutler, E. Three new variants of glucose-6-phosphate dehydrogenase associated with chronic nonspherocytic hemolytic anemia: G6PD Lincoln Park, G6PD Arlington Heights, and G6PD West Town. Am. J. Hemat. 6: 353-360, 1979. [PubMed: 532804] [Full Text: https://doi.org/10.1002/ajh.2830060407]

  135. Hook, E. B., Stamatoyannopoulos, G., Yoshida, A., Motulsky, A. G. Glucose-6-phosphate dehydrogenase Madrona: a slow electrophoretic glucose-6-phosphate dehydrogenase variant with kinetic characteristics similar to those of normal type. J. Lab. Clin. Med. 72: 404-409, 1968. [PubMed: 5674071]

  136. Hors-Cayla, M. C., Heuertz, S., Van Cong, N., Benne, F. Cattle gene mapping by somatic cell hybridization. (Abstract) Cytogenet. Cell Genet. 25: 165-166, 1979.

  137. Howell, E. B., Nelson, A. J., Jones, O. W. A new G-6-PD variant associated with chronic non-spherocytic haemolytic anaemia in a Negro family. J. Med. Genet. 9: 160-164, 1972. [PubMed: 5046624] [Full Text: https://doi.org/10.1136/jmg.9.2.160]

  138. Hutz, M. H., Yoshida, A., Salzano, F. M. Three rare G-6-PD variants from Porto Alegre, Brazil. Hum. Genet. 39: 191-197, 1977. [PubMed: 598829] [Full Text: https://doi.org/10.1007/BF00287011]

  139. Iancovici-Kidon, M., Sthoeger, D., Abrahamov, A., Wolach, B., Beutler, E., Gelbart, T., Barak, Y. A new exon 9 glucose-6-phosphate dehydrogenase mutation (G6PD 'Rehovot') in a Jewish Ethiopian family with variable phenotypes. Blood Cells Molec. Dis. 26: 567-571, 2000. Note: Erratum: Blood Cells Molec. Dis. 27: 351 only, 2001. [PubMed: 11112389] [Full Text: https://doi.org/10.1006/bcmd.2000.0334]

  140. Ishwad, C. S., Naik, S. N. A new glucose-6-phosphate dehydrogenase variant (G-6-PD Kalyan) found in a Koli family. Hum. Genet. 66: 171-175, 1984. [PubMed: 6714978] [Full Text: https://doi.org/10.1007/BF00286595]

  141. Iwai, K., Hirono, A., Matsuoka, H., Kawamoto, F., Horie, T., Lin, K., Tantular, I. S., Dachlan, Y. P., Notopuro, H., Hidayah, N. I., Salim, A. M. A., Fujii, H., Miwa, S., Ishii, A. Distribution of glucose-6-phosphate dehydrogenase mutations in Southeast Asia. Hum. Genet. 108: 445-449, 2001. [PubMed: 11499668] [Full Text: https://doi.org/10.1007/s004390100527]

  142. Jain, M., Cui, L., Brenner, D. A., Wang, B., Handy, D. E., Leopold, J. A., Loscalzo, J., Apstein, C. S., Liao, R. Increased myocardial dysfunction after ischemia-reperfusion in mice lacking glucose-6-phosphate dehydrogenase. Circulation 109: 898-903, 2004. [PubMed: 14757696] [Full Text: https://doi.org/10.1161/01.CIR.0000112605.43318.CA]

  143. Jiang, W., Yu, G., Liu, P., Geng, Q., Chen, L., Lin, Q., Ren, X., Ye, W., He, Y., Guo, Y., Duan, S., Wen, J., and 9 others. Structure and function of glucose-6-phosphate dehydrogenase-deficient variants in Chinese population. Hum. Genet. 119: 463-478, 2006. [PubMed: 16607506] [Full Text: https://doi.org/10.1007/s00439-005-0126-5]

  144. Johnson, G. J., Kaplan, M. E., Beutler, E. G6PD Long Prairie: a new mutant exhibiting normal sensitivity to inhibition by NADPH and accompanied by nonspherocytic hemolytic anemia. Blood 49: 247-251, 1977. [PubMed: 12846]

  145. Johnston, P. G., VandeBerg, J. L., Sharman, G. B. Inheritance of erythrocyte glucose 6-phosphate dehydrogenase in the red-necked wallaby, Macropus rufogriseus (Desmarest) consistent with paternal X inactivation. Biochem. Genet. 13: 235-242, 1975. [PubMed: 1147889] [Full Text: https://doi.org/10.1007/BF00486017]

  146. Junien, C., Kaplan, J. C., Meienhofer, M. C., Maigret, P., Sender, A. G6PD Baudelocque: a new unstable variant characterized in cultured fibroblasts. Enzyme 18: 45-59, 1974. [PubMed: 4852206]

  147. Kaeda, J. S., Chhotray, G. P., Ranjit, M. R., Bautista, J. M., Reddy, P. H., Stevens, D., Naidu, J. M., Britt, R. P., Vulliamy, T. J., Luzzatto, L., Mason, P. J. A new glucose-6-phosphate dehydrogenase variant, G6PD Orissa (44 ala-gly), is the major polymorphic variant in tribal populations in India. Am. J. Hum. Genet. 57: 1335-1341, 1995. [PubMed: 8533762]

  148. Kageoka, T., Satoh, C., Goriki, K., Fujita, M., Neriishi, S., Yamamura, K., Kaneko, J., Masunari, N. Electrophoretic variants of blood proteins in Japanese. IV. Prevalence and enzymologic characteristics of G6PD variants in Hiroshima and Nagasaki. Hum. Genet. 70: 101-108, 1985. [PubMed: 4007855] [Full Text: https://doi.org/10.1007/BF00273066]

  149. Kahn, A., Bernard, J.-F., Cottreau, D., Mazie, J., Boivin, P. Gd(-) Abrami, a deficient G6PD variant with hemizygous expression in blood cells of a woman with primary myelofibrosis. Humangenetik 30: 41-46, 1975. [PubMed: 241700] [Full Text: https://doi.org/10.1007/BF00273630]

  150. Kahn, A., Boivin, P., Hakim, J., Lagneau, J. Heterogeneite des glucose-6-phosphate dehydrogenase erythrocytaire deficitaires dans la race noire: etude cinetique et description de deux nouvelles variants Gd(-) Dakar et Gd(-) Mali. Nouv. Rev. Franc. Hemat. 11: 741-758, 1971. [PubMed: 4261190]

  151. Kahn, A., Boivin, P., Lagneau, J. Phenotypes de la glucose-6-phosphate dehydrogenase erythrocytaire dans la race noire. Humangenetik 18: 261-270, 1973. [PubMed: 4719636] [Full Text: https://doi.org/10.1007/BF00290606]

  152. Kahn, A., Boulard, M., Hakim, J., Schaison, G., Boivin, P., Bernard, J. Anemie hemolytique congenitale non spherocytaire par deficit en glucose 6-phosphate dehydrogenase erythrocytaire: description de deux nouvelles variants: Gd(-) Saint Louis (Paris) et Gd(-) Hayem. Nouv. Rev. Franc. Hemat. 14: 587-600, 1974. [PubMed: 4141762]

  153. Kahn, A., Dao, C., Cottreau, D., Bilski-Pasquier, G. 'Gd(-) Hotel Dieu': a new G-6PD variant with chronic hemolysis in a Negro patient from Senegal. Hum. Genet. 39: 353-358, 1977. [PubMed: 598844] [Full Text: https://doi.org/10.1007/BF00295431]

  154. Kahn, A., Esters, A., Habedank, M. Gd(-) Aachen, a new variant of deficient glucose-6-phosphate dehydrogenase. Hum. Genet. 32: 171-180, 1976. [PubMed: 1270075] [Full Text: https://doi.org/10.1007/BF00291500]

  155. Kahn, A., Hakim, J., Cottreau, D., Boivin, P. Gd (-) Matam. An African glucose 6-phosphate dehydrogenase variant with enzyme deficiency. Biochemical and immunological properties in various hemopoietic tissues. Clin. Chim. Acta 59: 183-190, 1975. [PubMed: 235389] [Full Text: https://doi.org/10.1016/0009-8981(75)90027-3]

  156. Kahn, A., North, M. L., Cottreau, D., Giron, G., Lang, J. M. G6PD Vientiane: a new glucose-6-phosphate dehydrogenase variant with increased stability. Hum. Genet. 43: 85-89, 1978. [PubMed: 27443] [Full Text: https://doi.org/10.1007/BF00396482]

  157. Kahn, A., North, M. L., Messer, J., Boivin, P. G-6PD 'Ankara': a new G-6PD variant with deficiency found in a Turkish family. Humangenetik 27: 247-250, 1975. [PubMed: 1150245] [Full Text: https://doi.org/10.1007/BF00278353]

  158. Kanno, H., Huang, I.-Y., Kan, Y. W., Yoshida, A. Two structural genes on different chromosomes are required for encoding the major subunit of human red cell glucose-6-phosphate dehydrogenase. Cell 58: 595-606, 1989. [PubMed: 2758468] [Full Text: https://doi.org/10.1016/0092-8674(89)90440-6]

  159. Kanno, H., Takano, T., Fujii, H., Tani, K., Morisaki, T., Hirono, A., Kumakawa, T., Ogura, H., Takahashi, K., Tsutsumi, H., Miwa, S. A new glucose-6-phosphate variant (G6PD Iwate) associated with congenital non-spherocytic hemolytic anemia. Acta Haemat. Jpn. 51: 715-719, 1987.

  160. Kaplan, J. C., Hanzlickova-Leroux, A., Nicholas, A. M., Rosa, R., Weiler, C., Lepercq, G. A new glucose-6-phosphate dehydrogenase variant (G6PD Port-Royal). Enzyme 12: 25-32, 1971. [PubMed: 5105366] [Full Text: https://doi.org/10.1159/000459511]

  161. Kaplan, J. C., Rosa, R., Seringe, P., Hoeffel, J. C. Le polymorphisme genetique de la glucose-6-phosphate deshydrogenase erythrocytaire chez l'homme. II. Etude d'une nouvelle variete a activite diminuee: le type 'Kabyle'. Enzymol. Biol. Clin. (Basel) 8: 332-340, 1967. [PubMed: 5299577]

  162. Kaplan, M., Hammerman, C., Vreman, H. J., Stevenson, D. K., Beutler, E. Acute hemolysis and severe neonatal hyperbilirubinemia in glucose-6-phosphate dehydrogenase deficient heterozygotes. J. Pediat. 139: 137-140, 2001. [PubMed: 11445808] [Full Text: https://doi.org/10.1067/mpd.2001.115312]

  163. Kaplan, M., Renbaum, P., Levy-Lahad, E., Hammerman, C., Lahad, A., Beutler, E. Gilbert syndrome and glucose-6-phosphate dehydrogenase deficiency: a dose-dependent genetic interaction crucial to neonatal hyperbilirubinemia. Proc. Nat. Acad. Sci. 94: 12128-12132, 1997. [PubMed: 9342374] [Full Text: https://doi.org/10.1073/pnas.94.22.12128]

  164. Karadsheh, N. S., Awidi, A. S., Tarawneh, M. S. Two new glucose-6 phosphate dehydrogenase (G6PD) variants associated with hemolytic anemia: G6PD Amman-1 and G6PD Amman-2. Am. J. Hemat. 22: 185-192, 1986. [PubMed: 3706292] [Full Text: https://doi.org/10.1002/ajh.2830220209]

  165. Kay, A. C., Kuhl, W., Prchal, J., Beutler, E. The origin of glucose-6-phosphate-dehydrogenase (G6PD) polymorphisms in African-Americans. Am. J. Hum. Genet. 50: 394-398, 1992. [PubMed: 1734718]

  166. Kirkman, H. N., Eng, L.-I. L. Variants of glucose 6-phosphate dehydrogenase in Indonesia. Nature 221: 959, 1969. [PubMed: 5765510] [Full Text: https://doi.org/10.1038/221959a0]

  167. Kirkman, H. N., Kidson, C., Kennedy, M. Variants of human glucose-6-phosphate dehydrogenase. Studies of samples from New Guinea. In: Beutler, E.: Hereditary Disorders of Erythrocyte Metabolism. New York: Grune and Stratton (pub.) 1968. Pp. 126-145.

  168. Kirkman, H. N., McCurdy, P. R., Naiman, J. L. Functionally abnormal glucose-6-phosphate dehydrogenases. Cold Spring Harbor Symp. Quant. Biol. 29: 391-398, 1964. [PubMed: 14278484] [Full Text: https://doi.org/10.1101/sqb.1964.029.01.041]

  169. Kirkman, H. N., Ramot, B., Lee, J. T. Altered aggregational properties in a genetic variant of human glucose-6-phosphate dehydrogenase. Biochem. Genet. 3: 137-150, 1969. [PubMed: 5364922] [Full Text: https://doi.org/10.1007/BF00520349]

  170. Kirkman, H. N., Riley, H. D., Jr. Congenital nonspherocytic hemolytic anemia. Am. J. Dis. Child. 102: 313-320, 1961. [PubMed: 13756265] [Full Text: https://doi.org/10.1001/archpedi.1961.02080010315006]

  171. Kirkman, H. N., Rosenthal, I. M., Simon, E. B., Carson, P. E., Brinson, A. G. 'Chicago I' variant of glucose-6-phosphate dehydrogenase in congenital hemolytic disease. J. Lab. Clin. Med. 63: 715-725, 1964. [PubMed: 14158057]

  172. Kirkman, H. N., Schettini, E., Pickard, B. M. Mediterranean variant of glucose-6-phosphate dehydrogenase. J. Lab. Clin. Med. 63: 726-735, 1964.

  173. Kirkman, H. N., Simon, E. R., Pickard, B. M. Seattle variant of glucose-6-phosphate dehydrogenase. J. Lab. Clin. Med. 66: 834-840, 1965. [PubMed: 5844610]

  174. Kissin, C., Cotte, J. Etude d'un variant de glucose-6-phosphate deshydrogenase: I B type Constantine. Enzymol. Biol. Clin. (Basel) 11: 277-284, 1970. [PubMed: 5414134] [Full Text: https://doi.org/10.1159/000458365]

  175. Kitao, T., Ito, K., Hattori, K., Matsuki, T., Yoneyama, Y. G6PD Kanazawa: a new variant of glucose-6-phosphate dehydrogenase associated with congenital nonspherocytic hemolytic anemia. Acta Haemat. 68: 131-135, 1982. [PubMed: 6812368] [Full Text: https://doi.org/10.1159/000206964]

  176. Kojima, H. Congenital nonspherocytic hemolytic disease (CNHD) due to a G-6-PD variant: G-6-PD Kyoto. Acta Haemat. Jpn. 35: 32-38, 1972. [PubMed: 4673853]

  177. Koliakos, G., Kalomenopoulou, M., Grammatikos, P., Dimitriadou, A., Kouzi-Koliakos, K., Zacharaki, R., Skaragas, G., Kokka, A., Trakatellis, A. A new glucose 6-phosphate dehydrogenase variant (G6PD Thessaloniki) in a patient with idiopathic myelofibrosis. Hum. Hered. 39: 141-149, 1989. [PubMed: 2591977] [Full Text: https://doi.org/10.1159/000153850]

  178. Krasnopolskaya, K. D., Bochkov, N. P. Genetic heterogeneity of hereditary enzymopathies. Vestn. Akad. Med. Nauk. SSSR 9: 56-64, 1982. [PubMed: 7148122]

  179. Krasnopolskaya, K. D., Shatskaya, T. L., Filippov, I. K., Annenkov, G. A., Zakharova, T. V., Mekhtiev, N. K., Movsum-Zade, K. M. Genetic heterogeneity of G6PD deficiency: study of mutant alleles in Shekii district of Azerbaijan. Genetika 13: 1455-1461, 1977. [PubMed: 144081]

  180. Kumakawa, T., Suzuki, S., Fujii, H., Miwa, S. Frequency of glucose 6-phosphate dehydrogenase (G6PD) deficiency in Tokyo and a new variant: G6PD Musashino. Acta Haemat. Jpn. 50: 25-28, 1987. [PubMed: 3591235]

  181. Kurdi-Haidar, B., Mason, P. J., Berrebi, A., Ankra-Badu, G., Al-Ali, A., Oppenheim, A., Luzzatto, L. Origin and spread of the glucose-6-phosphate dehydrogenase variant (G6PD-Mediterranean) in the Middle East. Am. J. Hum. Genet. 47: 1013-1019, 1990. [PubMed: 1978555]

  182. Kwiatkowska, J., Kacprzak-Bergman, I. New erythrocyte glucose 6-phosphate dehydrogenase variant. Acta Haemat. 46: 188-192, 1971. [PubMed: 4256637] [Full Text: https://doi.org/10.1159/000208573]

  183. Kwok, C. J., Martin, A. C. R., Au, S. W. N., Lam, V. M. S. G6PDdb, an integrated database of glucose-6-phosphate dehydrogenase (G6PD) mutations. Hum. Mutat. 19: 217-224, 2002. [PubMed: 11857737] [Full Text: https://doi.org/10.1002/humu.10036]

  184. Lenzerini, L., Meera Khan, P., Filippi, G., Rattazzi, M. C., Rat, A. K. Characterization of glucose-6-phosphate dehydrogenase variants. I. Occurrence of a G6PD Seattle-like variant in Sardinia and its interaction with G6PD Mediterranean variant. Am. J. Hum. Genet. 21: 142-153, 1969. [PubMed: 5305539]

  185. Leopold, J. A., Dam, A., Maron, B. A., Scribner, A. W., Liao, R., Handy, D. E., Stanton, R. C., Pitt, B., Loscalzo, J. Aldosterone impairs vascular reactivity by decreasing glucose-6-phosphate dehydrogenase activity. Nature Med. 13: 189-197, 2007. Note: Erratum: Nature Med. 15: 1093 only, 2009. [PubMed: 17273168] [Full Text: https://doi.org/10.1038/nm1545]

  186. Lisker, R., Linares, C., Motulsky, A. G. Glucose-6-phosphate dehydrogenase Mexico, a new variant with enzyme deficiency, abnormal mobility and absence of hemolysis. J. Lab. Clin. Med. 79: 788-793, 1972. [PubMed: 5018742]

  187. Lisker, R., Perez Briceno, R., Agrilar, L., Yoshida, A. A variant glucose-6-phosphate dehydrogenase Gd(-) Chiapas associated with moderate enzyme deficiency and occasional hemolytic anemia. Hum. Genet. 43: 81-84, 1978. [PubMed: 669721] [Full Text: https://doi.org/10.1007/BF00396481]

  188. Lisker, R., Perez Briceno, R., Zavala, C., Navarrette, J. I., Wessels, M., Yoshida, A. A glucose 6-phosphate dehydrogenase Gd(-) Castilla variant characterized by mild deficiency associated with drug-induced hemolytic anemia. J. Lab. Clin. Med. 90: 754-759, 1977. [PubMed: 903703]

  189. Lisker, R., Perez-Briceno, R., Beutler, E. A new glucose-6-phosphate dehydrogenase variant, Gd(-) Tepic, characterized by moderate enzyme deficiency and mild episodes of hemolytic anemia. Hum. Genet. 69: 19-21, 1985. [PubMed: 3967887] [Full Text: https://doi.org/10.1007/BF00295523]

  190. Lisker, R., Perez-Briceno, R., Rave, V., Yoshida, A. Glucosa-6-fosfato deshidrogenasa Gd(-) Distrito Federal: nueva variante asociada a deficiencia enzimatica moderada y anemia hemolitica ocasional. Rev. Invest. Clin. 33: 209-211, 1981. [PubMed: 7291768]

  191. Liu, Y., Phelan, J., Go, R. C. P., Prchal, J. F., Prchal, J. T. Rapid determination of clonality by detection of two closely-linked X chromosome exonic polymorphisms using allele-specific PCR. J. Clin. Invest. 99: 1984-1990, 1997. [PubMed: 9109443] [Full Text: https://doi.org/10.1172/JCI119366]

  192. Long, W. K., Kirkman, H. N., Sutton, H. H. Electrophoretically slow variants of glucose-6-phosphate dehydrogenase from red cells of Negroes. J. Lab. Clin. Med. 65: 81-87, 1965. [PubMed: 14260699]

  193. Longo, L., Vanegas, O. C., Patel, M., Rosti, V., Li, H., Waka, J., Merghoub, T., Pandolfi, P. P., Notaro, R., Manova, K., Luzzatto, L. Maternally transmitted severe glucose 6-phosphate dehydrogenase deficiency is an embryonic lethal. EMBO J. 21: 4229-4239, 2002. [PubMed: 12169625] [Full Text: https://doi.org/10.1093/emboj/cdf426]

  194. Louicharoen, C., Nuchprayoon, I. G6PD Viangchan (871G-A) is the most common G6PD-deficient variant in the Cambodian population. J. Hum. Genet. 50: 448-452, 2005. [PubMed: 16155737] [Full Text: https://doi.org/10.1007/s10038-005-0276-2]

  195. Louicharoen, C., Patin, E., Paul, R., Nuchprayoon, I., Witoonpanich, B., Peerapittayamongkol, C., Casademont, I., Sura, T., Laird, N. M., Singhasivanon, P., Quintana-Murci, L., Sakuntabhai, A. Positively selected G6PD-Mahidol mutation reduces Plasmodium vivax density in Southeast Asians. Science 326: 1546-1549, 2009. [PubMed: 20007901] [Full Text: https://doi.org/10.1126/science.1178849]

  196. Luzzatto, L., Afolayan, A. Enzyme properties of different types of human erythrocyte glucose-6-phosphate dehydrogenase with characterization of two new genetic variants. J. Clin. Invest. 47: 1833-1842, 1968. [PubMed: 5666113] [Full Text: https://doi.org/10.1172/JCI105873]

  197. Luzzatto, L., Usanga, E. A., Bienzle, U., Esan, G. F. J., Fusuan, F. A. Imbalance in X-chromosome expression: evidence for a human X-linked gene affecting growth of hemopoietic cells. Science 205: 1418-1420, 1979. [PubMed: 472761] [Full Text: https://doi.org/10.1126/science.472761]

  198. Luzzatto, L., Usanga, E. A., Reddy, S. Glucose-6-phosphate dehydrogenase deficient red cells: resistance to infection by malarial parasites. Science 164: 839-842, 1969. [PubMed: 4889647] [Full Text: https://doi.org/10.1126/science.164.3881.839]

  199. Luzzatto, L. Genetic heterogeneity and pathophysiology of G6PD deficiency. Brit. J. Haemat. 28: 151-156, 1974. [PubMed: 4154774] [Full Text: https://doi.org/10.1111/j.1365-2141.1974.tb06649.x]

  200. Luzzatto, L. Personal Communication. London, England 4/1990.

  201. MacDonald, D., Town, M., Mason, P., Vulliamy, T., Luzzatto, L., Goff, D. K. Deficiency in red blood cells. (Letter) Nature 350: 115, 1991. [PubMed: 2005960] [Full Text: https://doi.org/10.1038/350115a0]

  202. Maeda, M., Constantoulakis, P., Chen, C.-S., Stamatoyannopoulos, G., Yoshida, A. Molecular abnormalities of a human glucose-6-phosphate dehydrogenase variant associated with undetectable enzyme activity and immunologically cross-reacting material. Am. J. Hum. Genet. 51: 386-395, 1992. [PubMed: 1353664]

  203. Mamlok, R. J., Mamlok, V., Mills, G. C., Daeschner, C. W., III, Schmalstieg, F. C., Anderson, D. C. Glucose-6-phosphate dehydrogenase deficiency, neutrophil dysfunction and Chromobacterium violaceum sepsis. J. Pediat. 111: 852-854, 1987. [PubMed: 3681551] [Full Text: https://doi.org/10.1016/s0022-3476(87)80203-2]

  204. Mamlok, R. J., Mills, G. C., Goldblum, R. M., Daeschner, C. W. Glucose-6-phosphate dehydrogenase Beaumont: a new variant with severe enzyme deficiency and chronic nonspherocytic hemolytic anemia. Enzyme 34: 15-21, 1985. [PubMed: 4085453] [Full Text: https://doi.org/10.1159/000469354]

  205. Mandelli, F., Amadori, S., DeLaurenzi, A., Kahn, A., Isacchi, G., Papa, G. Glucose-6-phosphate dehydrogenase Velletri: a new variant with reduced activity in a patient with congenital non-spherocytic haemolytic anemia. Acta Haemat. 57: 121-126, 1977. [PubMed: 14470] [Full Text: https://doi.org/10.1159/000207869]

  206. Marks, P. A., Banks, J., Gross, R. Genetic heterogeneity of glucose-6-phosphate dehydrogenase deficiency. Nature 194: 454-456, 1962.

  207. Martin, S. K., Miller, L. H., Alling, D., Okoye, V. C., Esan, G. J. F., Osunkoya, B. O., Deane, M. Severe malaria and glucose-6-phosphate-dehydrogenase deficiency: a reappraisal of the malaria-G6PD hypothesis. Lancet 313: 524-526, 1979. Note: Originally Volume I. [PubMed: 85108] [Full Text: https://doi.org/10.1016/s0140-6736(79)90946-2]

  208. Martini, G., Toniolo, D., Vulliamy, T., Luzzatto, L., Dono, R., Viglietto, G., Paonessa, G., D'Urso, M., Persico, M. G. Structural analysis of the X-linked gene encoding human glucose 6-phosphate dehydrogenase. EMBO J. 5: 1849-1855, 1986. [PubMed: 2428611] [Full Text: https://doi.org/10.1002/j.1460-2075.1986.tb04436.x]

  209. Mason, P. J., Stevens, D. J., Luzzatto, L., Brenner, S., Aparicio, S. Genomic structure and sequence of the Fugu rubripes glucose-6-phosphate dehydrogenase gene (G6PD). Genomics 26: 587-591, 1995. [PubMed: 7607684] [Full Text: https://doi.org/10.1016/0888-7543(95)80179-p]

  210. Mason, P. J. New insights into G6PD deficiency. (Annotation) Brit. J. Haemat. 94: 585-591, 1996. [PubMed: 8826878]

  211. Matsuoka, H., Jichun, W., Hirai, M., Yoshida, S., Arai, M., Ishii, A., Baral, M. P. Two cases of glucose-6-phophate (sic) dehydrogenase-deficient Nepalese belonging to the G6PD Mediterranean-type, not India-Pakistan sub-type but Mediterranean-Middle East sub-type. J. Hum. Genet. 48: 275-277, 2003. [PubMed: 12768444] [Full Text: https://doi.org/10.1007/s10038-003-0018-2]

  212. Matsuoka, H., Nguon, C., Kanbe, T., Jalloh, A., Sato, H., Yoshida, S., Hirai, M., Arai, M., Socheat, D., Kawamoto, F. Glucose-6-phosphate dehydrogenase (G6PD) mutations in Cambodia: G6PD Viangchan (871G-A) is the most common variant in the Cambodian population. J. Hum. Genet. 50: 468-472, 2005. [PubMed: 16136268] [Full Text: https://doi.org/10.1007/s10038-005-0279-z]

  213. Matsuoka, H., Wang, J., Hirai, M., Arai, M., Yoshida, S., Kobayashi, T., Jalloh, A., Lin, K., Kawamoto, F. Glucose-6-phosphate dehydrogenase (G6PD) mutations in Myanmar: G6PD Mahidol (487G-A) is the most common variant in the Myanmar population. J. Hum. Genet. 49: 544-547, 2004. [PubMed: 15349799] [Full Text: https://doi.org/10.1007/s10038-004-0187-7]

  214. McCann, S. R., Smithwick, A. M., Temperley, I. J., Tipton, K. G6PD (Dublin): chronic non-spherocytic haemolytic anaemia resulting from glucose-6-phosphate dehydrogenase deficiency in an Irish kindred. J. Med. Genet. 17: 191-193, 1980. [PubMed: 7401130] [Full Text: https://doi.org/10.1136/jmg.17.3.191]

  215. McCurdy, P. R., Blackwell, R. Q., Todd, D., Tso, S. C., Tuchinda, S. Further studies on glucose-6-phosphate dehydrogenase deficiency in Chinese subjects. J. Lab. Clin. Med. 75: 788-797, 1970. [PubMed: 5439677]

  216. McCurdy, P. R., Kamel, K., Selim, O. Heterogeneity of red cell glucose 6-phosphate dehydrogenase (G6PD) deficiency in Egypt. J. Lab. Clin. Med. 84: 673-680, 1974. [PubMed: 4283789]

  217. McCurdy, P. R., Kirkman, H. N., Naiman, J. L., Jim, R. T. S., Pickard, B. M. A Chinese variant of glucose-6-phosphate dehydrogenase. J. Lab. Clin. Med. 67: 374-385, 1966. [PubMed: 4379606]

  218. McCurdy, P. R., Mahmood, L. Red cell glucose-6-phosphate dehydrogenase deficiency in Pakistan. J. Lab. Clin. Med. 76: 943-948, 1970. [PubMed: 5485383]

  219. McCurdy, P. R., Maldonado, N. I., Dillon, D. E., Conrad, M. E. Variants of glucose-6-phosphate dehydrogenase (G-6-PD) associated with G-6-PD deficiency in Puerto Ricans. J. Lab. Clin. Med. 82: 432-437, 1973. [PubMed: 4728291]

  220. McCurdy, P. R. Use of genetic linkage for the detection of female carriers of hemophilia. New Eng. J. Med. 285: 218-219, 1971. [PubMed: 5087725] [Full Text: https://doi.org/10.1056/NEJM197107222850409]

  221. Meloni, T., Carta, F., Forteleoni, G., Carta, A., Ena, F., Meloni, G. F. Glucose 6-phosphate dehydrogenase deficiency and cataract of patients in northern Sardinia. Am. J. Ophthal. 110: 661-664, 1990. [PubMed: 2248331] [Full Text: https://doi.org/10.1016/s0002-9394(14)77064-3]

  222. Mentzer, W. C., Jr., Warner, R., Addiego, J., Smith, B., Walter, T. G6PD San Francisco: a new variant of glucose-6-phosphate dehydrogenase associated with congenital nonspherocytic hemolytic anemia. Blood 55: 195-198, 1980. [PubMed: 7353069]

  223. Mesbah-Namin, S. A., Sanati, M. H., Mowjoodi, A., Mason, P. J., Vulliamy, T. J., Noori-Daloii, M. R. Three major glucose-6-phosphate dehydrogenase-deficient polymorphic variants identified in Mazandaran state of Iran. Brit. J. Haemat. 117: 763-764, 2002. [PubMed: 12028056] [Full Text: https://doi.org/10.1046/j.1365-2141.2002.03483.x]

  224. Miller, D. R., Wollman, M. R. A new variant of glucose 6-phosphate dehydrogenase deficiency hereditary hemolytic anemia, G6PD Cornell: erythrocyte, leukocyte and platelet studies. Blood 44: 323-331, 1974. [PubMed: 4152748]

  225. Mills, G. C., Alperin, J. B., Trimmer, K. B. Studies on variant glucose-6-phosphate dehydrogenase: G6PD Fort Worth. Biochem. Med. 13: 264-275, 1975. [PubMed: 1007] [Full Text: https://doi.org/10.1016/0006-2944(75)90084-8]

  226. Milner, G., Delamore, I. W., Yoshida, A. G-6-PD Manchester: a new variant associated with chronic nonspherocytic hemolytic anemia. Blood 43: 271-276, 1974. [PubMed: 4149233]

  227. Miwa, S., Fujii, H., Nakashima, K., Miura, Y., Yamada, K., Hagiwara, T., Fukuda, M. Three new electrophoretically normal glucose-6-phosphate dehydrogenase variants associated with congenital nonspherocytic hemolytic anemia found in Japan: G6PD Ogikubo, Yokohama, and Akita. Hum. Genet. 45: 11-17, 1978. [PubMed: 730178] [Full Text: https://doi.org/10.1007/BF00277568]

  228. Miwa, S., Fujii, H., Nakatsuji, T., Ishida, Y., Oda, E., Kaneto, A., Motokawa, M., Ariga, Y., Fukuchi, S., Sasai, S., Hiraoka, K., Kashii, H., Kodama, T. Four new electrophoretically slow-moving glucose-6-phosphate dehydrogenase variants associated with congenital nonspherocytic hemolytic anemia found in Japan: Gd(-) Kurume, Gd(-) Fukushima, Gd(-) Yamaguchi, and Gd(-) Wakayama. Am. J. Hemat. 5: 131-138, 1978. [PubMed: 736032] [Full Text: https://doi.org/10.1002/ajh.2830050208]

  229. Miwa, S., Fujii, H. Molecular basis of erythroenzymopathies associated with hereditary hemolytic anemia: tabulation of mutant enzymes. Am. J. Hemat. 51: 122-132, 1996. [PubMed: 8579052] [Full Text: https://doi.org/10.1002/(SICI)1096-8652(199602)51:2<122::AID-AJH5>3.0.CO;2-#]

  230. Miwa, S., Nakashima, K., Ono, J., Fujii, H., Suzuki, E. Three glucose 6-phosphate dehydrogenase variants found in Japan. Hum. Genet. 36: 327-334, 1977. [PubMed: 15940] [Full Text: https://doi.org/10.1007/BF00446284]

  231. Miwa, S., Ono, J., Nakashima, K., Abe, S., Kageoka, T., Shinohara, K., Isobe, J., Yamaguchi, H. Two new glucose 6-phosphate dehydrogenase variants associated with congenital nonspherocytic hemolytic anemia found in Japan: Gd(-) Tokushima and Gd(-) Tokyo. Am. J. Hemat. 1: 433-442, 1976. [PubMed: 1008056] [Full Text: https://doi.org/10.1002/ajh.2830010408]

  232. Modiano, G., Battistuzzi, G., Esan, G. J. F., Testa, U., Luzzatto, L. Genetic heterogeneity of 'normal' human erythrocyte glucose-6-phosphate dehydrogenase: an isoelectrophoretic polymorphism. Proc. Nat. Acad. Sci. 76: 852-856, 1979. [PubMed: 284409] [Full Text: https://doi.org/10.1073/pnas.76.2.852]

  233. Mohrenweiser, H. W., Neel, J. V. Frequency of thermostability variants: estimation of total 'rare' variant frequency in human populations. Proc. Nat. Acad. Sci. 78: 5729-5733, 1981. [PubMed: 6946512] [Full Text: https://doi.org/10.1073/pnas.78.9.5729]

  234. Morelli, A., Benatti, U., Guida, L., De Flora, A. G6PD Cagliari: a new low activity glucose 6-phosphate dehydrogenase variant characterized by enhanced intracellular lability. Hum. Genet. 66: 62-65, 1984. [PubMed: 6698555] [Full Text: https://doi.org/10.1007/BF00275188]

  235. Morisaki, T., Fujii, H., Takegawa, S., Tani, K., Hirono, A., Takizawa, T., Takahashi, K., Shinogi, M., Teshirogi, T., Miwa, S. G6PD Sendagi: a new glucose-6-phosphate dehydrogenase variant associated with congenital hemolytic anemia. Hum. Genet. 65: 214-215, 1983. [PubMed: 6654337] [Full Text: https://doi.org/10.1007/BF00286669]

  236. Nafa, K., Reghis, A., Osmani, N., Baghli, L., Benabadji, M., Kaplan, J.-C., Vulliamy, T. J., Luzzatto, L. G6PD Aures: a new mutation (48 ile-to-thr) causing mild G6PD deficiency is associated with favism. Hum. Molec. Genet. 2: 81-82, 1993. [PubMed: 8490627] [Full Text: https://doi.org/10.1093/hmg/2.1.81]

  237. Nagel, R. L., Ranney, H. M. Genetic epidemiology of structural mutations of the beta-globin gene. Semin. Hemat. 27: 342-359, 1990. [PubMed: 2255919]

  238. Nakai, T., Yoshida, A. G6PD Heian. A glucose-6-phosphate dehydrogenase variant associated with hemolytic anemia found in Japan. Clin. Chim. Acta 51: 199-203, 1974. [PubMed: 4828221] [Full Text: https://doi.org/10.1016/0009-8981(74)90030-8]

  239. Nakashima, K., Ono, J., Abe, S., Miwa, S., Yoshida, A. G6PD Ube: a glucose 6-phosphate dehydrogenase variant found in four unrelated Japanese families. Am. J. Hum. Genet. 29: 24-30, 1977. [PubMed: 835572]

  240. Nakatsuji, T., Miwa, S. Incidence and characteristics of glucose-6-phosphate dehydrogenase variants in Japan. Hum. Genet. 51: 297-305, 1979. [PubMed: 511159] [Full Text: https://doi.org/10.1007/BF00283398]

  241. Nance, W. E. Turner's syndrome, twinning, and an unusual variant of glucose-6-phosphate dehydrogenase. Am. J. Hum. Genet. 16: 380-392, 1964. [PubMed: 14207553]

  242. Necheles, T. F., Snyder, L. M., Strauss, W. Glucose-6-phosphate dehydrogenase Boston. A new variant associated with congenital nonspherocytic hemolytic disease. Humangenetik 13: 218-221, 1971. [PubMed: 4398877] [Full Text: https://doi.org/10.1007/BF00326945]

  243. Niazi, G., Adeyokunu, A., Westwood, B., Beutler, E. G6PD Aures: a rare mutant of G6PD in Saudi Arabia: molecular and clinical presentations. Saudi Med. J. 17: 311-314, 1996.

  244. Ninfali, P., Baronciani, L., Bardoni, A., Bresolin, N. Muscle expression of glucose-6-phosphate dehydrogenase deficiency in different variants. Clin. Genet. 48: 232-237, 1995. [PubMed: 8825599] [Full Text: https://doi.org/10.1111/j.1399-0004.1995.tb04095.x]

  245. Ninokata, A., Kimura, R., Samakkarn, U., Settheetham-Ishida, W., Ishida, T. Coexistence of five G6PD variants indicates ethnic complexity of Phuket islanders, Southern Thailand. J. Hum. Genet. 51: 424-428, 2006. [PubMed: 16528451] [Full Text: https://doi.org/10.1007/s10038-006-0380-y]

  246. Notaro, R., Afolayan, A., Luzzatto, L. Human mutations in glucose 6-phosphate dehydrogenase reflect evolutionary history. FASEB J. 14: 485-494, 2000. [PubMed: 10698963] [Full Text: https://doi.org/10.1096/fasebj.14.3.485]

  247. Nowicki, L., Strobel, S., Martin, H., Koschwitz, U. Ueber eine neue erythrocytaere glucose 6-phosphatdehydrogenase Variante, Typ Frankfurt. Klin. Wschr. 52: 478-484, 1974. [PubMed: 4408431] [Full Text: https://doi.org/10.1007/BF01468536]

  248. Nsouly, G. M., Prchal, J. T. Characterization of a new G6PD variant and its associated oxidative damage. Clin. Res. 29: 829, 1981.

  249. O'Brien, S. J. The extent and character of biochemical genetic variation in the domestic cat. J. Hered. 71: 2-8, 1980.

  250. Ogura, H., Morisaki, T., Tani, K., Kanno, H., Tsutsumi, H., Takahashi, K., Miyamori, T., Fujii, H., Miwa, S. A new glucose-6-phosphate dehydrogenase variant (G6PD Tsukui) associated with congenital hemolytic anemia. Hum. Genet. 78: 369-371, 1988. [PubMed: 3360447] [Full Text: https://doi.org/10.1007/BF00291738]

  251. Ohno, S. Sex Chromosomes and Sex-linked Genes. Berlin and New York: Springer (pub.) 1967.

  252. Othieno-Obel, A. East African variant of glucose-6-phosphate dehydrogenase. East Afr. Med. J. 49: 230-234, 1972. [PubMed: 5047808]

  253. Pai, G. S., Sprenkle, J. A., Do, T. T., Mareni, C. E., Migeon, B. R. Localization of loci for hypoxanthine phosphoribosyltransferase and glucose-6-phosphate dehydrogenase and biochemical evidence of nonrandom X chromosome expression from studies of a human X-autosome translocation. Proc. Nat. Acad. Sci. 77: 2810-2813, 1980. [PubMed: 6930669] [Full Text: https://doi.org/10.1073/pnas.77.5.2810]

  254. Panich, V., Bumrungtrakul, P., Jitjai, C., Kamolmatayakul, S., Khoprasert, B., Klaisuvan, C., Kongmuang, U., Maneechai, P., Pornpatkul, M., Ruengrairatanaroje, P., Surapruk, P., Viriyayudhakorn, S. Glucose-6-phosphate dehydrogenase deficiency in South Vietnamese. Hum. Hered. 30: 361-364, 1980. [PubMed: 7216225] [Full Text: https://doi.org/10.1159/000153158]

  255. Panich, V., Na-Nakorn, S. G6PD variants in Thailand. J. Med. Assoc. Thai. 63: 537-543, 1980. [PubMed: 7441068]

  256. Panich, V., Sungnate, T., Na-Nakorn, S. Acute intravascular hemolysis and renal failure in a new glucose 6-phosphate dehydrogenase variant: G6PD Siriraj. J. Med. Assoc. Thai. 55: 726-731, 1972. [PubMed: 4650733]

  257. Panich, V., Sungnate, T., Wasi, P., Na-Nakorn, S. G-6-PD Mahidol: the most common glucose-6-phosphate dehydrogenase variant in Thailand. J. Med. Assoc. Thai. 55: 576-585, 1972. [PubMed: 5081671]

  258. Panich, V., Sungnate, T. Characterization of glucose 6-phosphate dehydrogenase in Thailand: the occurrence of 6 variants among 50 G6PD deficient Thai. Humangenetik 18: 39-46, 1973. [PubMed: 4721339]

  259. Panich, V. G6PD characterization in Thailand. Genetics 74 (suppl.): s208 only, 1973.

  260. Panich, V. G6PD Intanon, a new glucose 6-phosphate dehydrogenase variant. Humangenetik 21: 203-205, 1974. [PubMed: 4837298] [Full Text: https://doi.org/10.1007/BF00281039]

  261. Panich, V. G6PD variants in Laotians. Hum. Hered. 24: 285-290, 1974. [PubMed: 4435794] [Full Text: https://doi.org/10.1159/000152661]

  262. Panich, V. Glucose-6-phosphate dehydrogenase in Thailand. Hum. Genet. 53: 227-228, 1980. [PubMed: 7358391] [Full Text: https://doi.org/10.1007/BF00273501]

  263. Pawlak, A. L., Mazurkiewicz, C. A., Ordynski, J., Ruzynkowa, D., Horst, A. G6PD Poznan, variant with severe enzyme deficiency. Humangenetik 28: 163-165, 1975. [PubMed: 1150271] [Full Text: https://doi.org/10.1007/BF00735750]

  264. Pawlak, A. L., Zagorski, Z., Rozynkowa, D., Horst, A. Polish variant of glucose-6-phosphate dehydrogenase (G-6-PD Lublin). Humangenetik 10: 340-343, 1970. [PubMed: 5493238] [Full Text: https://doi.org/10.1007/BF00278771]

  265. Pekrun, A., Eber, S. W., Schroter, W. G6PD Avenches and G6PD Moosburg: biochemical and erythrocyte membrane characterization. Blut 58: 11-14, 1989. [PubMed: 2917200] [Full Text: https://doi.org/10.1007/BF00320229]

  266. Perroni, L., Tassara, P., Baldi, M., Reali, R., Scartezzini, P. G6PD variants detected in Genoa area. In: Weatherall, D. J.; Fiorelli, G.; Gorini, S.: Advances in Red Blood Cell Biology. New York: Raven Press (pub.) 1982. Pp. 409-416.

  267. Persico, M. G., Toniolo, D., Nobile, C., D'Urso, M., Luzzatto, L. cDNA sequences of human glucose 6-phosphate dehydrogenase cloned in pBR322. Nature 294: 778-780, 1981. [PubMed: 6895658] [Full Text: https://doi.org/10.1038/294778a0]

  268. Persico, M. G., Viglietto, G., Martini, G., Toniolo, D., Paonessa, G., Moscatelli, C., Dono, R., Vulliamy, T., Luzzatto, L., D'Urso, M. Isolation of human glucose-6-phosphate dehydrogenase (G6PD) cDNA clones: primary structure of the protein and unusual 5-prime non-coding region. Nucleic Acids Res. 14: 2511-2522, 1986. Note: Erratum: Nucleic Acids Res. 14: 7822 only, 1986. [PubMed: 3515319] [Full Text: https://doi.org/10.1093/nar/14.6.2511]

  269. Picat, C., Etiemble, J., Boivin, P., Le Prise, P.-Y. Gd(-) Rennes, a new deficient variant of glucose-6-phosphate dehydrogenase associated with congenital nonspherocytic hemolytic anemia found in France. Hum. Genet. 55: 125-127, 1980. [PubMed: 7450748] [Full Text: https://doi.org/10.1007/BF00329139]

  270. Pinto, P. V. C., Newton, W. A., Jr., Richardson, K. E. Evidence for four types of erythrocyte glucose-6-phosphate dehydrogenase from G-6-PD deficient human subjects. J. Clin. Invest. 45: 823-831, 1966. [PubMed: 5913292] [Full Text: https://doi.org/10.1172/JCI105398]

  271. Poggi, V. Personal Communication. London, England 1989.

  272. Poon, M.-C., Hall, K., Scott, C. W., Prchal, J. T. G6PD Viangchan: a new glucose 6-phosphate dehydrogenase variant from Laos. Hum. Genet. 78: 98-99, 1988. [PubMed: 3338798] [Full Text: https://doi.org/10.1007/BF00291246]

  273. Porter, I. H., Boyer, S. H., Watson-Williams, E. J., Adam, A., Szeinberg, A., Siniscalco, M. Variation of glucose-6-phosphate dehydrogenase in different populations. Lancet 283: 895-899, 1964. Note: Originally Volume I. [PubMed: 14124079] [Full Text: https://doi.org/10.1016/s0140-6736(64)91626-5]

  274. Porter, I. H., Schulze, J., McKusick, V. A. Genetical linkage between the loci for glucose-6-phosphate dehydrogenase deficiency and colour-blindness in American Negroes. Ann. Hum. Genet. 26: 107-122, 1962. [PubMed: 13985567] [Full Text: https://doi.org/10.1111/j.1469-1809.1962.tb01316.x]

  275. Prchal, J., Moreno, H., Conrad, M., Vitek, A. G-6-PD Dothan: a new variant associated with chronic hemolytic anemia. I.R.C.S. 7: 348, 1979.

  276. Prchal, J. T., Crist, W. M., Malluh, A., Vitek, A., Tauxe, W. N., Carroll, A. J. A new glucose-6-phosphate dehydrogenase deficient variant in a patient with Chediak-Higashi syndrome. Blood 56: 476-480, 1980. [PubMed: 7407414]

  277. Prchal, J. T., Hall, K., Csepreghy, M., Lilly, M., Berkow, R., Scott, C. W. Two apparent glucose-6-phosphate dehydrogenase variants in normal XY males: G6PD Alabama. Am. J. Med. 84: 517-523, 1988. [PubMed: 3348252] [Full Text: https://doi.org/10.1016/0002-9343(88)90276-8]

  278. Ramot, B., Ben-Bassat, I., Shchory, M. New glucose-6-phosphate dehydrogenase variants observed in Israel and their association with congenital nonspherocytic hemolytic disease. J. Lab. Clin. Med. 74: 895-901, 1969. [PubMed: 5359663]

  279. Ramot, B., Brok, F. A new glucose-6-phosphate dehydrogenase mutant (Tel-Hashomer mutant). Ann. Hum. Genet. 28: 167-172, 1964. [PubMed: 14228003] [Full Text: https://doi.org/10.1111/j.1469-1809.1964.tb00471.x]

  280. Rattazzi, M. C., Corash, L. M., Van Zzanen, G. E., Jaffe, E. R., Piomelli, S. G6PD deficiency and chronic hemolysis: four new mutants--relationships between clinical syndrome and enzyme kinetics. Blood 38: 205-218, 1971. [PubMed: 4397482]

  281. Rattazzi, M. C., Lenzerini, L., Meera Khan, P., Luzzatto, L. Characterization of glucose-6-phosphate dehydrogenase variants. II. G6PD Kephalonia, G6PD Attica, and G6PD 'Seattle-like' found in Greece. Am. J. Hum. Genet. 21: 154-167, 1969. [PubMed: 5770172]

  282. Ravindranath, Y., Beutler, E. Two new variants of glucose-6-phosphate dehydrogenase associated with hereditary non-spherocytic hemolytic anemia: G6PD Wayne and G6PD Huron. Am. J. Hemat. 24: 357-363, 1987. [PubMed: 3565372] [Full Text: https://doi.org/10.1002/ajh.2830240405]

  283. Reys, L., Manso, C., Stamatoyannopoulos, G. Genetic studies on Southeastern Bantu of Mozambique. I. Variants of glucose-6-phosphate dehydrogenase. Am. J. Hum. Genet. 22: 203-215, 1970. [PubMed: 5435642]

  284. Rosenstraus, M., Chasin, L. A. Isolation of mammalian cell mutants deficient in glucose-6-phosphate dehydrogenase activity: linkage to hypoxanthine phosphoribosyl transferase. Proc. Nat. Acad. Sci. 72: 493-497, 1975. [PubMed: 1054832] [Full Text: https://doi.org/10.1073/pnas.72.2.493]

  285. Roth, E. F., Jr., Raventos-Suarez, C., Rinaldi, A., Nagel, R. L. Glucose-6-phosphate dehydrogenase deficiency inhibits in vitro growth of Plasmodium falciparum. Proc. Nat. Acad. Sci. 80: 298-299, 1983. [PubMed: 6337374] [Full Text: https://doi.org/10.1073/pnas.80.1.298]

  286. Roychoudhury, A. K., Nei, M. Human Polymorphic Genes: World Distribution. New York: Oxford Univ. Press (pub.) 1988.

  287. Ruwende, C., Khoo, S. C., Snow, R. W., Yates, S. N. R., Kwiatkoweld, D., Gupta, S., Warn, P., Alisopp, G. E. M., Gilbert, S. C., Peschu, N., Newbold, C. I., Greenwood, S. M., Marsh, K., Hill, A. V. S. Natural selection of hemi- and heterozygotes for G6PD deficiency in Africa by resistance to severe malaria. Nature 376: 246-249, 1995. [PubMed: 7617034] [Full Text: https://doi.org/10.1038/376246a0]

  288. Saenz, G. F., Chaves, M., Berrantes, A., Elizondo, J., Montero, A. G., Yoshida, A. A glucose-6-phosphate dehydrogenase variant, Gd(-) Santamaria found in Costa Rica. Acta Haemat. 72: 37-40, 1984. [PubMed: 6433630] [Full Text: https://doi.org/10.1159/000206354]

  289. Saidi, N., Hors-Cayla, M. C., Van Cong, N., Benne, F. Sheep gene mapping by somatic cell hybridization. (Abstract) Cytogenet. Cell Genet. 25: 200, 1979.

  290. Samuel, A. P. W., Saha, N., Omer, A., Hoffbrand, A. V. Quantitative expression of G6PD activity of different phenotypes of G6PD and haemoglobin in a Sudanese population. Hum. Hered. 31: 110-115, 1981. [PubMed: 7228057] [Full Text: https://doi.org/10.1159/000153188]

  291. Sansone, G., Perroni, L., Testa, U., Mareni, C., Luzzatto, L. New genetic variants of glucose 6-phosphate dehydrogenase (G6PD) in Italy. Ann. Hum. Genet. 45: 97-104, 1981. [PubMed: 7316485] [Full Text: https://doi.org/10.1111/j.1469-1809.1981.tb00310.x]

  292. Sansone, G., Perroni, L., Yoshida, A., Dave, V. A new glucose 6-phosphate dehydrogenase variant (Gd Trinacria) in two unrelated families of Sicilian ancestry. Ital. J. Biochem. 26: 44-50, 1977. [PubMed: 16832]

  293. Sansone, G., Perroni, L., Yoshida, A. Glucose-6-phosphate dehydrogenase variants from Italian subjects associated with severe neonatal jaundice. Brit. J. Haemat. 31: 159-165, 1975. [PubMed: 1201235] [Full Text: https://doi.org/10.1111/j.1365-2141.1975.tb00846.x]

  294. Shatskaya, T. L., Krasnopolskaya, K. D., Annenkov, G. A. A description of new mutant forms of erythrocyte glucose-6-phosphate dehydrogenase isolated at the territory of the Soviet Union. Genetika 11: 116-122, 1975.

  295. Shatskaya, T. L., Krasnopolskaya, K. D., Idelson, L. J. Mutant forms of erythrocyte glucose 6-phosphate dehydrogenase in Ashkenazi: description of two new variants, G6PD Kirovograd and G6PD Zhitomir. Hum. Genet. 33: 175-178, 1976. [PubMed: 133080] [Full Text: https://doi.org/10.1007/BF00281892]

  296. Shatskaya, T. L., Krasnopolskaya, K. D., Idelson, L. J. The new form of glucose 6-phosphate dehydrogenase (G6PD 'Kaluga') from erythrocytes of a patient with chronic non-spherocytic hemolytic anemia. Vopr. Med. Khim. 22: 764-768, 1976. [PubMed: 17221]

  297. Shatskaya, T. L., Krasnopolskaya, K. D., Tzoneva, M., Mavrudieva, M., Toncheva, D. Variants of erythrocyte glucose-6-phosphate dehydrogenase (G6PD) in Bulgarian populations. Hum. Genet. 54: 115-117, 1980. [PubMed: 7390473] [Full Text: https://doi.org/10.1007/BF00279060]

  298. Shatskaya, T. L., Krasnopolskaya, K. D., Zakharova, T. V. Regularities of distribution of Gd-alleles in Azerbaijan. II. Identification of G6PD mutant forms. Genetika 16: 2217-2225, 1980.

  299. Shows, T. B., Brown, J. A., Chapman, V. M. Comparative gene mapping of HPRT, G6PD and PGK in man, mouse, and Muntjac deer. Birth Defects Orig. Art. Ser. XII(7): 436-439, 1976. [PubMed: 1024651]

  300. Shows, T. B., Brown, J. A. Human X-linked genes regionally mapped utilizing X-autosome translocations and somatic cell hybrids. Proc. Nat. Acad. Sci. 72: 2125-2129, 1975. [PubMed: 1056018] [Full Text: https://doi.org/10.1073/pnas.72.6.2125]

  301. Shows, T. B., Tashian, R. E., Brewer, G. J. Erythrocyte glucose-6-phosphate dehydrogenase in Caucasians: new inherited variant. Science 145: 1056-1057, 1964. [PubMed: 14172618] [Full Text: https://doi.org/10.1126/science.145.3636.1056]

  302. Sidi, Y., Aderka, D., Brok-Simoni, F., Benjamin, D., Ramot, B., Pinkhas, J. Viral hepatitis with extreme hyperbilirubinemia, massive hemolysis and encephalopathy in a patient with a new G6PD variant. Isr. J. Med. Sci. 16: 130-133, 1980. [PubMed: 7364565]

  303. Siegel, N. H., Beutler, E. Hemolytic anemia caused by G-6-PD Carswell, a new variant. Ann. Intern. Med. 75: 437-439, 1971. [PubMed: 5568155] [Full Text: https://doi.org/10.7326/0003-4819-75-3-437]

  304. Smith, J. E., Ryer, K., Wallace, L. Glucose-6-phosphate dehydrogenase deficiency in a dog. Enzyme 21: 379-382, 1976. [PubMed: 939229] [Full Text: https://doi.org/10.1159/000458882]

  305. Snyder, L. M., Necheles, T. F., Reddy, W. J. G-6-PD Worcester: a new variant, associated with X-linked optic atrophy. Am. J. Med. 49: 125-132, 1970. [PubMed: 5311120] [Full Text: https://doi.org/10.1016/s0002-9343(70)80122-x]

  306. Stamatoyannopoulos, G., Kotsakis, P., Voigtlander, V., Akrivakis, A., Motulsky, A. G. Electrophoretic diversity of glucose-6-phosphate dehydrogenase among Greeks. Am. J. Hum. Genet. 22: 587-596, 1970. [PubMed: 5516240]

  307. Stamatoyannopoulos, G., Voigtlander, V., Akrivakis, A. Thessaly variant of glucose-6-phosphate dehydrogenase. Humangenetik 9: 23-25, 1970. [PubMed: 5496124] [Full Text: https://doi.org/10.1007/BF00696009]

  308. Stamatoyannopoulos, G., Voigtlander, V., Kotsakis, P., Akrivakis, A. Genetic diversity of the 'Mediterranean' glucose-6-phosphate dehydrogenase deficiency phenotype. J. Clin. Invest. 50: 1253-1261, 1971. [PubMed: 4397065] [Full Text: https://doi.org/10.1172/JCI106603]

  309. Stamatoyannopoulos, G., Yoshida, A., Bacopoulos, C., Motulsky, A. G. Athens variant of glucose-6-phosphate dehydrogenase. Science 157: 831-833, 1967. [PubMed: 17842795] [Full Text: https://doi.org/10.1126/science.157.3790.831]

  310. Stevens, D. J., Wanachiwanawin, W., Mason, P. J., Vulliamy, T. J., Luzzatto, L. G6PD Canton a common deficient variant in South East Asia caused by a 459 arg-to-leu mutation. Nucleic Acids Res. 18: 7190, 1990. [PubMed: 2263506] [Full Text: https://doi.org/10.1093/nar/18.23.7190]

  311. Stocco dos Santos, R. C., Barretto, O. C. O., Nonoyama, K., Castro, N. H. C., Ferraz, O. P., Walter-Moura, J., Vescio, C. C. S., Becak, W. X-linked syndrome: mental retardation, hip luxation, and G6PD variant (Gd(+) Butantan). Am. J. Med. Genet. 39: 133-136, 1991. [PubMed: 2063914] [Full Text: https://doi.org/10.1002/ajmg.1320390204]

  312. Stocco dos Santos, R. C., Castro, N. H. C., Holmes, A. L., Becak, W., Tackels-Horne, D., Lindsey, C. J., Lubs, H. A., Stevenson, R. E., Schwartz, C. E. Stocco dos Santos X-linked mental retardation syndrome: clinical elucidation and localization to Xp11.3-Xq21.3. Am. J. Med. Genet. 118A: 255-259, 2003. [PubMed: 12673656] [Full Text: https://doi.org/10.1002/ajmg.a.20021]

  313. Streiff, F., Vigneron, C. Anemie hemolytique chronique par deficit en glucose 6-phosphate deshydrogenase dans une famille d'origine Lorraine. Nouv. Rev. Franc. Hemat. 11: 279-290, 1971. [PubMed: 4252680]

  314. Takahashi, K., Fujii, H., Takegawa, S., Tani, K., Hirono, A., Takizawa, T., Kawakatsu, T., Miwa, S. A new glucose-6-phosphate dehydrogenase variant (G6PD Nagano) associated with congenital hemolytic anemia. Hum. Genet. 62: 368-370, 1982. [PubMed: 7166314] [Full Text: https://doi.org/10.1007/BF00304560]

  315. Takizawa, T., Fujii, H., Takegawa, S., Takahashi, K., Hirono, A., Morisaki, T., Kanno, H., Oka, R., Yoshioka, H., Miwa, S. A unique electrophoretic slow-moving glucose 6-phosphate dehydrogenase variant (G6PD Asahikawa) with a markedly acidic pH optimum. Hum. Genet. 68: 70-72, 1984. [PubMed: 6500558] [Full Text: https://doi.org/10.1007/BF00293876]

  316. Takizawa, T., Huang, I.-Y., Ikuta, T., Yoshida, A. Human glucose-6-phosphate dehydrogenase: primary structure and cDNA cloning. Proc. Nat. Acad. Sci. 83: 4157-4161, 1986. [PubMed: 3012556] [Full Text: https://doi.org/10.1073/pnas.83.12.4157]

  317. Takizawa, T., Yoneyama, Y., Miwa, S., Yoshida, A. A single nucleotide base transition is the basis of the common human glucose-6-phosphate dehydrogenase variant A(+). Genomics 1: 228-231, 1987. [PubMed: 3446582] [Full Text: https://doi.org/10.1016/0888-7543(87)90048-6]

  318. Takizawa, T., Yoshida, A. Molecular abnormality of the common glucose-6-phosphate dehydrogenase variant, G6PD A(+), and restriction-fragment-length polymorphism. (Abstract) Am. J. Hum. Genet. 41: A241, 1987.

  319. Talalak, P., Beutler, E. G-6-PD Bangkok: a new variant found in congenital nonspherocytic hemolytic disease (CNHD). Blood 33: 772-776, 1969. [PubMed: 5779160]

  320. Tanaka, K. R., Beutler, E. Hereditary hemolytic anemia due to glucose-6-phosphate dehydrogenase Torrance: a new variant. J. Lab. Clin. Med. 73: 657-667, 1969. [PubMed: 4388268]

  321. Tang, T. K., Huang, C.-S., Huang, M. J., Tam, K.-B., Yeh, C.-H., Tang, C.-J. C. Diverse point mutations result in glucose-6-phosphate dehydrogenase (G6PD) polymorphism in Taiwan. Blood 79: 2135-2140, 1992. [PubMed: 1562739]

  322. Testa, U., Meloni, T., Lania, A., Battistuzzi, G., Cutillo, S., Luzzatto, L. Genetic heterogeneity of glucose-6-phosphate dehydrogenase deficiency in Sardinia. Hum. Genet. 56: 99-105, 1980. [PubMed: 7203486] [Full Text: https://doi.org/10.1007/BF00281577]

  323. Thigpen, J. T., Steinberg, M. H., Beutler, E., Gillespie, G. T., Jr., Dreiling, B. J., Morrison, F. S. Glucose-6-phosphate dehydrogenase Jackson, a new variant associated with hemolytic anemia. Acta Haemat. 51: 310-314, 1974. [PubMed: 4209318] [Full Text: https://doi.org/10.1159/000208311]

  324. Tishkoff, S. A., Varkonyi, R., Cahinhinan, N., Abbes, S., Argyropoulos, G., Destro-Bisol, G., Drousiotou, A., Dangerfield, B., Lefranc, G., Loiselet, J., Piro, A., Stoneking, M., Tagarelli, A., Tagarelli, G., Touma, E. H., Williams, S. M., Clark, A. G. Haplotype diversity and linkage disequilibrium at human G6PD: recent origin of alleles that confer malarial resistance. Science 293: 455-462, 2001. [PubMed: 11423617] [Full Text: https://doi.org/10.1126/science.1061573]

  325. Tokarev, Y. N., Chernyak, N. B., Batischev, A. I., Lanzina, N. V., Alexeyev, G. A. Etude des proprietes electrophoretiques et cinetiques de la glucose-6-phosphate deshydrogenase (Gd) d'erythrocytes dans les deficits hereditaires de l'enzyme: description d'une nouvelle variante de glucose-6-phosphate deshydrogenase: la Gd Kremenchug. Nouv. Rev. Franc. Hemat. 20: 557-564, 1978. [PubMed: 752149]

  326. Toncheva, D., Tzoneva, M. Genetic polymorphism of G6PD in a Bulgarian population. Hum. Genet. 67: 340-342, 1984. [PubMed: 6540752] [Full Text: https://doi.org/10.1007/BF00291365]

  327. Toncheva, D. Variants of glucose-6-phosphate dehydrogenase in a Vietnamese population. Hum. Hered. 36: 348-351, 1986. [PubMed: 3793116] [Full Text: https://doi.org/10.1159/000153658]

  328. Toniolo, D., Martini, G., Migeon, B. R., Dono, R. Expression of the G6PD locus on the human X chromosome is associated with demethylation of three CpG islands within 100 kb of DNA. EMBO J. 7: 401-406, 1988. [PubMed: 2835227] [Full Text: https://doi.org/10.1002/j.1460-2075.1988.tb02827.x]

  329. Town, M., Athanasiou-Metaxa, M., Luzzatto, L. Intragenic interspecific complementation of glucose 6-phosphate dehydrogenase in human-hamster cell hybrids. Somat. Cell Molec. Genet. 16: 97-108, 1990. [PubMed: 2157298] [Full Text: https://doi.org/10.1007/BF01233040]

  330. Town, M., Bautista, J. M., Mason, P. J., Luzzatto, L. Both mutations in G6PD A- are necessary to produce the G6PD deficient phenotype. Hum. Molec. Genet. 1: 171-174, 1992. [PubMed: 1303173] [Full Text: https://doi.org/10.1093/hmg/1.3.171]

  331. Usanga, E. A., Bienzle, U., Cancedda, K., Fasuan, F. A., Ajayi, O., Luzzatto, L. Genetic variants of human erythrocyte glucose 6-phosphate dehydrogenase: new variants in West Africa characterized by column chromatography. Ann. Hum. Genet. 40: 279-286, 1977. [PubMed: 848857] [Full Text: https://doi.org/10.1111/j.1469-1809.1977.tb00192.x]

  332. Vaca, G., Ibarra, B., Garcia Cruz, D., Medina, C., Romero, F., Cantu, J. M., Beutler, E. G-6-PD Jalisco and G-6-PD Morelia: two new Mexican variants. Hum. Genet. 71: 82-85, 1985. [PubMed: 4029955] [Full Text: https://doi.org/10.1007/BF00295674]

  333. Vaca, G., Ibarra, B., Romero, F., Olivares, N., Cantu, J. M., Beutler, E. G-6-PD Guadalajara: a new mutant associated with chronic nonspherocytic hemolytic anemia. Hum. Genet. 61: 175-176, 1982. [PubMed: 7129446] [Full Text: https://doi.org/10.1007/BF00274214]

  334. van Bruggen, R., Bautista, J. M., Petropoulou, T., de Boer, M., van Zwieten, R., Gomez-Gallego, F., Belohradsky, B. H., Hartwig, N. G., Stevens, D., Mason, P. J., Roos, D. Deletion of leucine 61 in glucose-6-phosphate dehydrogenase leads to chronic nonspherocytic anemia, granulocyte dysfunction, and increased susceptibility to infections. Blood 100: 1026-1030, 2002. [PubMed: 12130518] [Full Text: https://doi.org/10.1182/blood.v100.3.1026]

  335. Ventura, A., Panizon, F., Soranzo, M. R., Veneziano, G., Sansone, G., Testa, U., Luzzatto, L. Congenital dyserythropoietic anaemia type II associated with a new type of G6PD deficiency (G6PD Gabrovizza). Acta Haemat. 71: 227-234, 1984. [PubMed: 6426235] [Full Text: https://doi.org/10.1159/000206592]

  336. Vergnes, H., Gherardi, M., Bouloux, C. Erythrocyte glucose-6-phosphate dehydrogenase in the Niokolonko (Malinke of the Niokolo) of the Eastern Senegal: identification of a slow variant with normal activity (Tacoma-like). Hum. Hered. 25: 80-87, 1975. [PubMed: 1150303] [Full Text: https://doi.org/10.1159/000152711]

  337. Vergnes, H., Gherardi, M., Quilici, J. C., Yoshida, A., Giacardy, R. G6PD Luz-Saint-Sauveur: a new variant with abnormal electrophoretic mobility, mild enzyme deficiency and absence of hemolytic disorders. I.R.C.S. 7: 14, 1973.

  338. Vergnes, H., Gherardi, M., Yoshida, A. G6PD Lozere and Trinacria-like: segregation of two non-hemolytic variants in a French family. Hum. Genet. 34: 293-298, 1976. [PubMed: 1002152] [Full Text: https://doi.org/10.1007/BF00295293]

  339. Vergnes, H., Ribet, A., Bommelaer, G., Amadieu, J., Brun, H. GD(-) Muret and GD(-) Colomiers, two new variants of glucose-6-phosphate dehydrogenase associated with favism. Hum. Genet. 57: 332-334, 1981. [PubMed: 7250973] [Full Text: https://doi.org/10.1007/BF00278958]

  340. Vergnes, H., Yoshida, A., Gourdin, D., Gherardi, M., Bierme, R., Ruffie, J. Glucose 6-phosphate dehydrogenase Toulouse: a new variant with marked instability and severe deficiency discovered in a family of Mediterranean ancestry. Acta Haemat. 51: 240-249, 1974. [PubMed: 4152602] [Full Text: https://doi.org/10.1159/000208300]

  341. Viglietto, G., Montanaro, V., Calabro, V., Vallone, D., D'Urso, M., Persico, M. G., Battistuzzi, G. Common glucose-6-phosphate dehydrogenase (G6PD) variants from the Italian population: biochemical and molecular characterization. Ann. Hum. Genet. 54: 1-15, 1990. [PubMed: 2321910] [Full Text: https://doi.org/10.1111/j.1469-1809.1990.tb00355.x]

  342. Vives-Corrons, J. L., Feliu, E., Pujades, M. A., Cardellach, F., Rozman, C., Carreras, A., Jou, J. M., Vallespi, M. T., Zuazu, F. J. Severe glucose-6-phosphate dehydrogenase (G6PD) deficiency associated with chronic hemolytic anemia, granulocyte dysfunction, and increased susceptibility to infections: description of a new molecular variant (G6PD Barcelona). Blood 59: 428-434, 1982. [PubMed: 7055648]

  343. Vives-Corrons, J. L., Pujades, A., Curia, M. D. Caracterizacion molecular de la glucosa-6-fosfato deshidrogenasa (G6PD) en 24 casos de deficit enzimatico y descripcion de una nueva variante (G6PD-Betica). Sangre 25: 1049-1064, 1980. [PubMed: 7221827]

  344. Vives-Corrons, J. L., Pujades, A. Heterogeneity of 'Mediterranean type' glucose-6-phosphate dehydrogenase (G6PD) deficiency in Spain and description of two new variants associated with favism. Hum. Genet. 60: 216-221, 1982. [PubMed: 7106752] [Full Text: https://doi.org/10.1007/BF00303006]

  345. Vives-Corrons, J. L., Pujades, M. A., Petit, J., Colomer, D., Corbella, M., Aguilar i Bascompte, J. L., Merino, A. Chronic nonspherocytic hemolytic anemia (CNSHA) and glucose 6 phosphate dehydrogenase (G6PD) deficiency in a patient with familial amyloidotic polyneuropathy (FAP): molecular study of a new variant (G6PD Clinic) with markedly acidic pH optimum. Hum. Genet. 81: 161-164, 1989. [PubMed: 2912886] [Full Text: https://doi.org/10.1007/BF00293894]

  346. Vives-Corrons, J.-L., Kuhl, W., Pujades, M. A., Beutler, E. Molecular genetics of the glucose-6-phosphate dehydrogenase (G6PD) Mediterranean variant and description of a new G6PD mutant, G6PD Andalus(1361A). Am. J. Hum. Genet. 47: 575-579, 1990. [PubMed: 2393028]

  347. Vulliamy, T., Beutler, E., Luzzatto, L. Variants of glucose-6-phosphate dehydrogenase are due to missense mutations spread throughout the coding region of the gene. Hum. Mutat. 2: 159-167, 1993. [PubMed: 8364584] [Full Text: https://doi.org/10.1002/humu.1380020302]

  348. Vulliamy, T. J., D'Urso, M., Battistuzzi, G., Estrada, M., Foulkes, N. S., Martini, G., Calabro, V., Poggi, V., Giordano, R., Town, M., Luzzatto, L., Persico, M. G. Diverse point mutations in the human glucose-6-phosphate dehydrogenase gene cause enzyme deficiency and mild or severe hemolytic anemia. Proc. Nat. Acad. Sci. 85: 5171-5175, 1988. [PubMed: 3393536] [Full Text: https://doi.org/10.1073/pnas.85.14.5171]

  349. Vulliamy, T. J., Kaeda, J. S., Ait-Chafa, D., Mangerini, R., Roper, D., Barbot, J., Mehta, A. B., Athanassiou-Metaxa, M., Luzzatto, L., Mason, P. J. Clinical and haematological consequences of recurrent G6PD mutations and a single new mutation causing chronic nonspherocytic haemolytic anaemia. Brit. J. Haemat. 101: 670-675, 1998. [PubMed: 9674740] [Full Text: https://doi.org/10.1046/j.1365-2141.1998.00763.x]

  350. Vulliamy, T. J., Othman, A., Town, M., Nathwani, A., Falusi, A. G., Mason, P. J., Luzzatto, L. Polymorphic sites in the African population detected by sequence analysis of the glucose-6-phosphate dehydrogenase gene outline the evolution of the variants A and A-. Proc. Nat. Acad. Sci. 88: 8568-8571, 1991. [PubMed: 1924316] [Full Text: https://doi.org/10.1073/pnas.88.19.8568]

  351. Vulliamy, T. J., Wanachiwanawin, W., Mason, P. J., Luzzatto, L. G6PD Mahidol, a common deficient variant in South East Asia is caused by a (163)glycine-to-serine mutation. Nucleic Acids Res. 17: 5868, 1989. [PubMed: 2503817] [Full Text: https://doi.org/10.1093/nar/17.14.5868]

  352. Vulliamy, T. J. Personal Communication. London, England 1989.

  353. Vulliamy, T., Mason, P., Luzzatto, L. The molecular basis of glucose-6-phosphate dehydrogenase deficiency. Trends Genet. 8: 138-143, 1992. [PubMed: 1631957] [Full Text: https://doi.org/10.1016/0168-9525(92)90372-B]

  354. Vulliamy, T., Rovira, A., Yusoff, N., Colomer, D., Luzzatto, L., Vives-Corrons, J.-L. Independent origin of single and double mutations in the human glucose 6-phosphate dehydrogenase gene. Hum. Mutat. 8: 311-318, 1996. [PubMed: 8956035] [Full Text: https://doi.org/10.1002/(SICI)1098-1004(1996)8:4<311::AID-HUMU3>3.0.CO;2-A]

  355. Vuopio, P., Harkonen, M., Helske, T., Naeveri, H. Red cell glucose-6-phosphate dehydrogenase deficiency in Finland: characterization of a new variant with severe enzyme deficiency. Scand. J. Haemat. 15: 145-152, 1975. [PubMed: 1188317] [Full Text: https://doi.org/10.1111/j.1600-0609.1975.tb01066.x]

  356. Vuopio, P., Harkonen, M., Johnsson, P., Nuutinen, M. Red cell glucose-phosphate dehydrogenase deficiency in Finland. Ann. Clin. Res. 5: 168-178, 1973. [PubMed: 4749241]

  357. Waitz, R., Boivin, P., Oberling, F., Casenave, J. P., North, M. L., Mayer, S. Variante Gd(-) Strasbourg de la glucose-6-phosphate dehydrogenase. Nouv. Rev. Franc. Hemat. 10: 312-314, 1970. [PubMed: 4246285]

  358. Weimer, T. A., Salzano, F. M., Hutz, M. H. Erythrocyte isozymes and hemoglobin types in a southern Brazilian population. J. Hum. Evol. 10: 319-328, 1981.

  359. Weimer, T. A., Schuler, L., Beutler, E., Salzano, F. M. Gd(+) Laguna, a new rare glucose-6-phosphate dehydrogenase variant from Brazil. Hum. Genet. 65: 402-404, 1984. [PubMed: 6693129] [Full Text: https://doi.org/10.1007/BF00291568]

  360. Weinreich, J., Busch, D., Gottstein, U., Schaefer, J., Rohr, J. Ueber zwei neue Faelle von hereditaerer nichtsphaerocytaerer haemolytischer Anaemie bei glucose-6-phosphat-dehydrogenase-Defekt in einer Nord Deutschen Familie. Klin. Wschr. 46: 146-149, 1968. [PubMed: 5667020] [Full Text: https://doi.org/10.1007/BF01727373]

  361. Welch, S. G., McGregor, I. A., Williams, K. A new variant of human erythrocyte G6PD occurring at a high frequency amongst the population of two villages in The Gambia, West Africa. Hum. Genet. 40: 305-309, 1978. [PubMed: 631849] [Full Text: https://doi.org/10.1007/BF00272191]

  362. Westring, D. W., Pisciotta, A. V. Anemia, cataracts, and seizures in patients with glucose-6-phosphate dehydrogenase deficiency. Arch. Intern. Med. 118: 385-390, 1966. [PubMed: 5925237]

  363. WHO. Nomenclature of glucose-6-phosphate dehydrogenase in man. Bull. World Health Organ. 36: 319-322, 1967. Note: See Also: Canad. Med. Assoc. J. 97: 422-424, 1967. [PubMed: 5299754]

  364. WHO. Standardization of procedures for the study of glucose-6-phosphate dehydrogenase: report of a WHO Scientific Group. World Health Organ. Tech. Rep. Ser. 366: 1-53, 1967. [PubMed: 4963040]

  365. Wilson, W. W. Congenital hemolytic anemia due to a deficiency of glucose 6-phosphate dehydrogenase. Rocky Mt. Med. J. 73: 160-162, 1976.

  366. Witt, I., Yoshioka, S. Biochemical characterization of a glucose-6-phosphate dehydrogenase variant with favism: G-6-PD Zaehringen. Klin. Wschr. 50: 205-209, 1972. [PubMed: 5011553] [Full Text: https://doi.org/10.1007/BF01487145]

  367. Wong, P. W. K., Shih, L.-Y., Hsia, D. Y. Y. Characterization of glucose-6-phosphate dehydrogenase among Chinese. Nature 208: 1323-1324, 1965. [PubMed: 5870191] [Full Text: https://doi.org/10.1038/2081323a0]

  368. Yermakov, N., Tokarev, J., Chernjak, N., Schoenian, G., Grieger, M., Guckler, G., Jacobasch, G., Mahmudova, M., Bahramov, S. New stable mutant Gd(-) variants: G6PD Tashkent and G6PD Nucus: molecular basis of hereditary enzyme deficiency. Acta Biol. Med. Ger. 40: 559-562, 1981. [PubMed: 7315103]

  369. Yoshida, A., Baur, E. W., Motulsky, A. G. A Philippino glucose-6-phosphate dehydrogenase variant (G6PD Union) with enzyme deficiency and altered substrate specificity. Blood 35: 506-513, 1970. [PubMed: 4392654]

  370. Yoshida, A., Beutler, E., Motulsky, A. G. Table of human glucose-6-phosphate dehydrogenase variants. Bull. WHO 45: 243-253, 1971. [PubMed: 5316621]

  371. Yoshida, A., Beutler, E. Human glucose-6-phosphate dehydrogenase variants: a supplementary tabulation. Ann. Hum. Genet. 41: 347-355, 1978. [PubMed: 626479] [Full Text: https://doi.org/10.1111/j.1469-1809.1978.tb01902.x]

  372. Yoshida, A., Stamatoyannopoulos, G., Motulsky, A. G. Negro variant of glucose-6-phosphate dehydrogenase deficiency (A-) in man. Science 155: 97-99, 1967. [PubMed: 6015571] [Full Text: https://doi.org/10.1126/science.155.3758.97]

  373. Yoshida, A., Takizawa, T., Prchal, J. T. RFLP of the X chromosome-linked glucose-6-phosphate dehydrogenase locus in blacks. Am. J. Hum. Genet. 42: 872-876, 1988. [PubMed: 2897162]

  374. Yoshida, A., Takizawa, T. The same extra FokI cleavage site exists in glucose-6-phosphate dehydrogenase variants A(+) and A(-). Am. J. Hum. Genet. 43: 131-133, 1988. [PubMed: 2840819]

  375. Yoshida, A. A single amino acid substitution (asparagine to aspartic acid) between normal (B plus) and the common Negro variant (A plus) of human glucose-6-phosphate dehydrogenase. Proc. Nat. Acad. Sci. 57: 835-840, 1967. [PubMed: 16591538] [Full Text: https://doi.org/10.1073/pnas.57.3.835]

  376. Yoshida, A. Human glucose-6-phosphate dehydrogenase: purification and characterization of Negro type variant (A+) and comparison with normal enzyme (B+). Biochem. Genet. 1: 81-99, 1967. [PubMed: 4388132] [Full Text: https://doi.org/10.1007/BF00486511]

  377. Yoshida, A. Amino acid substitution (histidine to tyrosine) in a glucose-6-phosphate dehydrogenase variant (G6PD Hektoen) associated with over-production. J. Molec. Biol. 52: 483-490, 1970. [PubMed: 5492291] [Full Text: https://doi.org/10.1016/0022-2836(70)90414-6]

  378. Yoshida, A. Personal Communication. Duarte, Calif. 2/26/1996.

  379. Zuo, L., Chen, E., Du, C. S., Chang, C. N., Chiu, D. T. Y. Genetic study of Chinese G6PD variants by direct PCR sequencing. (Abstract) Blood 76 (suppl. 1): 51A, 1990.


Contributors:
Carol A. Bocchini - updated : 10/24/2013
Patricia A. Hartz - updated : 7/11/2013
Ada Hamosh - updated : 1/6/2010
Cassandra L. Kniffin - updated : 2/12/2009
Marla J. F. O'Neill - updated : 11/17/2008
Cassandra L. Kniffin - updated : 1/14/2008
Marla J. F. O'Neill - updated : 4/12/2007
Cassandra L. Kniffin - updated : 7/14/2006
Cassandra L. Kniffin - updated : 6/26/2006
Cassandra L. Kniffin - updated : 3/9/2006
Marla J. F. O'Neill - updated : 1/18/2006
Victor A. McKusick - updated : 12/16/2005
Victor A. McKusick - updated : 1/11/2005
Victor A. McKusick - updated : 1/3/2005
Victor A. McKusick - updated : 8/19/2003
Victor A. McKusick - updated : 4/24/2003
Victor A. McKusick - updated : 3/4/2003
Victor A. McKusick - updated : 1/14/2003
Victor A. McKusick - updated : 10/22/2002
Victor A. McKusick - updated : 9/12/2002
Victor A. McKusick - updated : 4/23/2002
Victor A. McKusick - updated : 4/4/2002
Deborah L. Stone - updated : 10/8/2001
Ada Hamosh - updated : 9/24/2001
Ada Hamosh - updated : 8/27/2001
Victor A. McKusick - updated : 7/13/2000
Victor A. McKusick - updated : 4/26/2000
Victor A. McKusick - updated : 3/15/1999
Victor A. McKusick - updated : 11/12/1998
Victor A. McKusick - updated : 10/13/1998
Victor A. McKusick - updated : 6/25/1998
Victor A. McKusick - updated : 3/24/1998
Michael J. Wright - updated : 9/25/1997
Victor A. McKusick - updated : 9/19/1997
Victor A. McKusick - updated : 3/18/1997
Moyra Smith - updated : 11/12/1996
Alan F. Scott - updated : 12/14/1995

Creation Date:
Victor A. McKusick : 7/6/1987

Edit History:
carol : 09/05/2023
carol : 08/30/2023
alopez : 08/09/2023
alopez : 04/28/2023
carol : 11/21/2022
carol : 04/01/2022
carol : 03/25/2022
carol : 03/04/2022
carol : 08/05/2019
carol : 06/20/2019
carol : 05/11/2018
carol : 05/24/2017
carol : 11/08/2016
carol : 09/12/2016
carol : 09/09/2016
carol : 07/28/2016
carol : 06/05/2015
alopez : 5/22/2015
alopez : 5/21/2015
carol : 9/4/2014
carol : 8/30/2014
carol : 6/18/2014
carol : 3/11/2014
joanna : 12/9/2013
mgross : 10/25/2013
carol : 10/25/2013
carol : 10/24/2013
carol : 10/24/2013
carol : 10/24/2013
mgross : 7/11/2013
carol : 4/18/2013
terry : 3/14/2013
terry : 12/20/2012
alopez : 3/5/2012
mgross : 7/7/2011
carol : 4/7/2011
terry : 11/8/2010
terry : 10/12/2010
alopez : 1/19/2010
terry : 1/6/2010
terry : 6/12/2009
terry : 3/27/2009
terry : 3/27/2009
wwang : 2/19/2009
ckniffin : 2/12/2009
wwang : 11/24/2008
terry : 11/21/2008
terry : 11/17/2008
wwang : 1/31/2008
ckniffin : 1/14/2008
wwang : 4/18/2007
terry : 4/12/2007
alopez : 2/1/2007
carol : 7/21/2006
carol : 7/21/2006
ckniffin : 7/14/2006
wwang : 7/13/2006
ckniffin : 6/26/2006
wwang : 5/25/2006
ckniffin : 3/9/2006
wwang : 1/18/2006
wwang : 12/28/2005
terry : 12/16/2005
terry : 2/7/2005
tkritzer : 1/21/2005
terry : 1/11/2005
wwang : 1/6/2005
terry : 1/3/2005
terry : 5/21/2004
joanna : 3/17/2004
carol : 2/23/2004
cwells : 11/7/2003
cwells : 11/7/2003
mgross : 8/21/2003
terry : 8/19/2003
carol : 4/24/2003
terry : 4/24/2003
cwells : 3/11/2003
terry : 3/4/2003
carol : 1/22/2003
tkritzer : 1/17/2003
terry : 1/14/2003
tkritzer : 10/29/2002
tkritzer : 10/24/2002
terry : 10/22/2002
carol : 9/18/2002
tkritzer : 9/13/2002
tkritzer : 9/13/2002
tkritzer : 9/12/2002
tkritzer : 9/12/2002
carol : 7/2/2002
alopez : 5/3/2002
terry : 4/23/2002
cwells : 4/15/2002
cwells : 4/9/2002
terry : 4/4/2002
carol : 10/8/2001
alopez : 9/25/2001
terry : 9/24/2001
alopez : 8/30/2001
terry : 8/27/2001
alopez : 7/21/2000
terry : 7/13/2000
mcapotos : 5/24/2000
mcapotos : 5/19/2000
terry : 4/26/2000
carol : 9/10/1999
joanna : 6/18/1999
carol : 5/27/1999
carol : 3/18/1999
terry : 3/15/1999
carol : 11/19/1998
terry : 11/12/1998
carol : 10/20/1998
carol : 10/18/1998
terry : 10/13/1998
dkim : 7/21/1998
alopez : 6/26/1998
terry : 6/25/1998
alopez : 3/31/1998
terry : 3/24/1998
dholmes : 2/10/1998
alopez : 1/14/1998
terry : 1/12/1998
terry : 1/12/1998
dholmes : 1/9/1998
alopez : 11/10/1997
alopez : 11/10/1997
alopez : 11/10/1997
terry : 9/19/1997
alopez : 7/28/1997
terry : 7/9/1997
mark : 7/7/1997
alopez : 6/27/1997
alopez : 6/27/1997
alopez : 6/27/1997
alopez : 6/27/1997
alopez : 6/27/1997
joanna : 6/23/1997
carol : 6/23/1997
mark : 6/16/1997
mark : 3/18/1997
terry : 3/11/1997
terry : 1/27/1997
jamie : 1/21/1997
terry : 1/14/1997
joanna : 1/1/1997
terry : 11/19/1996
mark : 11/12/1996
mark : 9/18/1996
mark : 7/9/1996
mark : 3/31/1996
terry : 3/21/1996
joanna : 3/15/1996
mark : 3/1/1996
terry : 3/1/1996
mark : 2/19/1996
terry : 2/15/1996
terry : 2/6/1996
mark : 1/17/1996
mark : 1/17/1996
joanna : 12/18/1995
mark : 12/15/1995
terry : 12/14/1995
mark : 7/19/1995
carol : 2/27/1995
davew : 8/5/1994
jason : 7/1/1994
pfoster : 4/22/1994
terry : 4/20/1994