Entry - *120920 - CD46 ANTIGEN; CD46 - OMIM
 
* 120920

CD46 ANTIGEN; CD46


Alternative titles; symbols

TROPHOBLAST-LYMPHOCYTE CROSS-REACTIVE ANTIGEN; TLX
MEMBRANE COFACTOR PROTEIN; MCP
COMPLEMENT MEMBRANE COFACTOR PROTEIN
ANTIGEN DEFINED BY MONOCLONAL ANTIBODY TRA-2-10; MIC10


HGNC Approved Gene Symbol: CD46

Cytogenetic location: 1q32.2     Genomic coordinates (GRCh38): 1:207,752,038-207,795,516 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
1q32.2 {Hemolytic uremic syndrome, atypical, susceptibility to, 2} 612922 AD, AR 3

TEXT

Description

MCP, a C3B/C4B-binding molecule of the complement system with cofactor activity for the I-dependent cleavage of C3B and C4B, is widely distributed in white blood cells, platelets, epithelial cells, and fibroblasts (Lublin et al., 1988).


Cloning and Expression

Lublin et al. (1988) purified MCP from a human T-cell line and determined the sequence of the N-terminal 24 amino acids. An oligonucleotide probe was used to identify a clone from a human monocyte cDNA library. The deduced full-length MCP consists of a 34-amino acid signal peptide and a 350-amino acid mature protein. The protein has, beginning at the N terminus, 4 cysteine-rich repeating units (short consensus repeats, or SCRs) of about 60 amino acids each that match the consensus sequence found in a multigene family of complement regulatory proteins: CR1 (120620), CR2 (120650), C4BP (120830), CFH (134370), and DAF (125240). Immediately C-terminal of the SCRs is a serine/threonine/proline (STP)-rich region, a likely area for extensive O-glycosylation. MCP also has a transmembrane domain, a basic amino acid anchor, and a cytoplasmic tail.

Purcell et al. (1991) and Post et al. (1991) identified 4 and 6 isoforms of MCP, respectively. Post et al. (1991) demonstrated that the 6 isoforms vary in having 1 of 2 cytoplasmic tails and by having either all 3 STP regions (termed A, B, and C) or only STP-BC or STP-C. They showed that the STP-C isoforms are expressed as 45- to 55-kD proteins, the STP-BC isoforms are expressed as 55- to 65-kD proteins, and the STP-ABC isoforms are expressed as 65- to 75-kD proteins. The 65- to 75-kD variants were not expressed on peripheral blood cells or cell lines. Post et al. (1991) concluded that the presence of the B region of the STP area, which is richer in O-linked sugars, determines the expression of the 2 broad protein species. They also noted that up to 4 different forms of MCP are expressed on a single cell.


Gene Function

McIntyre et al. (1983) found that antisera to human syncytiotrophoblast microvillus cell surface membranes from different placentas are cytotoxic for lymphocytes from some persons but not others. Study of 10 antisera on lymphocytes from 30 donors suggested the presence of 3 distinct TLX groupings. McIntyre et al. (1983) proposed that TLX alloantigens are central in establishing maternal recognition and protection of the blastocyst, and that lack of recognition results in implantation failure and spontaneous abortion.

Dorig et al. (1993) used a genetic approach to identify the receptor for measles virus (MV). They tested human/rodent somatic cell hybrids for their ability to bind the Edmonston strain of MV and found that only cells containing human chromosome 1 were capable of binding virus. Rodent cells could not bind MV. A study of lymphocyte markers suggested that CD46 is the MV receptor. Dorig et al. (1993) showed that hamster cell lines expressing human CD46 could bind MV. Furthermore, infected CD46+ cells produced syncytia and viral proteins. Finally, polyclonal antisera against CD46 inhibited MV binding and infection. Dorig et al. (1993) concluded that CD46 is receptor for the Edmonston strain of MV. However, Tatsuo et al. (2000) noted that although the Edmonston strain of MV and the vaccine strains derived from it use CD46 as a cellular receptor, most clinical isolates do not. They showed that SLAM (603492), but not CD46, could act as a cellular receptor for clinical strains of MV.

Human herpesvirus-6 (HHV-6) is the etiologic agent of exanthema subitum, causes opportunistic infections in immunocompromised patients, and has been implicated in multiple sclerosis and in the progression of AIDS. Santoro et al. (1999) showed that the 2 major HHV-6 subgroups (A and B) use human CD46 as a cellular receptor. Downregulation of surface CD46 was documented during the course of HHV-6 infection. Both acute infection and cell fusion mediated by HHV-6 were specifically inhibited by a monoclonal antibody to CD46; fusion was also blocked by soluble CD46. Nonhuman cells that were resistant to HHV-6 fusion and entry became susceptible upon expression of recombinant human CD46.

CD46 acts as a cellular receptor for type IV pili of pathogenic Neisseria. The binding of piliated bacteria to host cells can be inhibited by antibody to CD46 or the recombinant molecule. Kallstrom et al. (2001) showed that the BC1 phenotype of the STP domain as well as the complement control protein repeat 3 (CCP3) of CD46 are important for efficient adherence of N. gonorrhoeae to host cells.

Kemper et al. (2003) examined the requirements for activation of T-regulatory-1 (Tr1) cells, which are defined as CD4 (186940)-positive T lymphocytes that secrete IL10 (124092) and suppress T-helper cells. Stimulation of purified CD4-positive T cells with monoclonal antibodies to CD3 (see 186740) and CD46 in the presence of IL2 (147680) or anti-CD28 (186760) induced the secretion of large amounts of IL10 and sustained proliferation, as measured by flow cytometric analysis for expression of PCNA (176740). CD45RA-positive/CD45RO-negative (naive) T cells and CD45RA-positive/CD45RO-positive (high-responding) T cells produced IL10 in response to these agonists, while CD45RA-negative/CD45RO-positive (memory) T cells did not. After primary anti-CD3/anti-CD46 activation, however, both naive and high-responding CD4-positive T cells acquired an IL10-producing memory phenotype (CD45RA-negative/CD45RO-positive). Stimulation of CD4-positive T cells with anti-CD3/anti-CD28 without anti-CD46 failed to induce IL10 production and caused the production of large amounts of IL2. Stimulation with anti-CD3/anti-CD28 in the presence of complement factor C3b (120700) dimers resulted in IL10 secretion comparable to that of anti-CD3/anti-CD28/anti-CD46-activated T cells. Supernatants of the anti-CD3/anti-CD46-activated T cells induced IL10-mediated suppression of proliferation by bystander T cells. Kemper et al. (2003) concluded that CD46 has a role in human T-cell regulation and that these findings establish a link between the complement system and adaptive immunity. They proposed that Tr1 cells are essential for maintaining peripheral tolerance and preventing autoimmunity, as well as for responses to many pathogens.

Kallstrom et al. (1997) identified CD46 as a human cell surface receptor for piliated pathogenic Neisseria. Johansson et al. (2003) generated transgenic mice expressing human CD46 and found they were susceptible to meningococcal disease because bacteria crossed the blood-brain barrier in these mice. Development of disease was more efficient with piliated bacteria after intranasal but not intraperitoneal challenge of Cd46 transgenic mice, suggesting that human CD46 facilitates pilus-dependent interactions at the epithelial mucosa.

Gaggar et al. (2003) showed that, unlike most adenoviruses, group B adenoviruses use CD46 rather than CAR (CXADR; 602621) to infect cells. Mass spectrometric, immunoblot, and fluorescence microscopy analyses determined that the group B fiber knob domain interacts with CD46. The authors found that, with the exception of Ad3, viruses of the B1 respiratory and B2 kidney and urinary tract subgroups both use the CD46 receptor. Expression of human CD46 rendered nonhuman cells susceptible to group B adenovirus infection in vitro and in vivo, and infection could be blocked by CD46 siRNA or soluble CD46 protein. Gaggar et al. (2003) concluded that CD46 is an essential receptor for group B adenoviruses and that this knowledge may facilitate the development of novel methods for adenovirus-mediated gene transfer.

The placenta is an immunologically privileged site. Using DNA microarrays to compare gene expression patterns, Sood et al. (2006) found that 3 regulators of complement, CD55 (125240), CD59 (107271), and MCP, are expressed at higher levels in normal placental villus sections compared with other normal human tissues. Within the placenta, CD55 and CD59 are expressed at greatest levels in amnion, followed by chorion and villus sections, whereas MCP is expressed at higher levels only in villus sections. These inhibitors of complement are expressed on syncytiotrophoblasts, the specialized placental cells lining the villi that are in direct contact with maternal blood. The amnion compared with chorion is remarkably nonimmunogenic, and the immune properties of the amnion are intriguing because it is not in direct contact with maternal cells. Sood et al. (2006) suggested that the amnion may secrete the complement inhibitors themselves or in the form of protected exosomes into the amniotic fluid or the neighboring maternofetal junction.

Using confocal microscopy, Oliaro et al. (2006) demonstrated that treatment of T or natural killer (NK) cytotoxic lymphocytes with anti-CD46 beads, but not anti-transferrin receptor (TFRC; 190010) beads, resulted in recruitment of the microtubule-organizing center (MTOC) and perforin (PRF1; 170280) to the site of CD46 ligation, indicating cell polarization. Ligation with soluble anti-CD46 altered T-cell polarization, inhibiting IFNG (147570) production. CD46 ligation also prevented normal polarization of T cells towards antigen-presenting cells (APCs) and prevented immune synapse formation, but it did not affect interaction of NK cells with target cells. Cells expressing measles hemagglutinin ligated CD46 on T cells and recruited the MTOC and CD3 to the site. These T cells only produced IFNG with the addition of anti-CD3/CD28. Oliaro et al. (2006) concluded that external signals can alter lymphocyte polarization toward APC or target cells and inhibit lymphocyte function.


Gene Structure

By genomic sequence analysis, Post et al. (1991) determined that the MCP gene contains 14 exons and spans 43 kb. Exons 7, 8, and 9 encode the A, B, and C regions of the STP area, respectively. Exon 13 encodes a 16-residue cytoplasmic tail, whereas use of exon 14 results in a 23-amino acid tail.

Cui et al. (1993) analyzed the promoter region of the CD46 gene. They detected a GC-rich region as well as a CAAT box in reverse orientation surrounded by 4 putative SP1-binding sites, but no TATA element, suggesting that CD46 is a housekeeping gene. Reporter assays determined that promoter activity is confined to the GC-rich region.


Mapping

Andrews et al. (1985) studied an antigen expressed by most human cells, but not erythrocytes, and defined by monoclonal antibody TRA-2-10. The antigen was expressed on the surface of human-mouse somatic cell hybrids; segregation analysis showed that the antigen is determined by a gene (MCP1) on human chromosome 1.

Lublin et al. (1988) localized the MCP gene to 1q31-q41 by Southern analysis of human-rodent somatic cell hybrid DNA and by in situ hybridization. This was the sixth member of this multigene family that had been assigned to this region of the genome. Bora et al. (1989) demonstrated that the MCP gene is on the same 1,250-kb NotI fragment that contains CR1, CR2, DAF, and C4BP and maps within 100 kb of the 3-prime end of the CR1 gene. The order of the genes appears to be that just indicated, with MCP preceding the other 4 genes.


Molecular Genetics

Atypical Hemolytic Uremic Syndrome, Susceptibility to, 2

Noris et al. (2003) identified a heterozygous mutation (120920.0001) in the MCP gene in 2 patients with a family history of atypical hemolytic uremic syndrome (AHUS2; 612922). The mutation caused a change in 3 amino acids at position 233-235 and insertion of a premature stop codon, which resulted in loss of the transmembrane domain of the protein and severely reduced cell-surface expression of MCP.

Like factor H (CFH; 134370), MCP inhibits complement activation by regulating C3b deposition on targets. Richards et al. (2003) hypothesized that MCP mutations could predispose to aHUS, and they sequenced MCP coding exons in affected members from 30 affected families. They identified mutations in the MCP gene in affected members from 3 of these families: a heterozygous 6-bp deletion in one family (120920.0002) and a ser206-to-pro mutation (S206P; 120920.0003), which was heterozygous in one family and homozygous in another. An individual with the 6-bp deletion had reduced MCP levels and approximately 50% C3b binding compared with normal controls. Individuals with the S206P mutation expressed normal quantities of protein, but demonstrated approximately 50% reduction in C3b binding in heterozygotes and complete lack of C3b binding in homozygotes. Studies in transfectants showed that the deletion mutant was retained intracellularly. S206P protein was expressed on the cell surface but had a reduced ability to prevent complement activation, consistent with its reduced C3b binding and cofactor activity.

Esparza-Gordillo et al. (2005) identified a specific SNP haplotype in the MCP gene, which was overrepresented in aHUS patients and strongly associated with the severity of the disease. Linkage disequilibrium analysis suggested that the haplotype included the CR1 (120620), DAF (CD55; 125240), and C4BPA (120830) genes. Two SNPs in the haplotype influenced the transcription activity of the MCP promoter in transient transfection experiments. The SNP haplotype block was particularly frequent among patients who carried mutations in HF1 (CFH; 134370), MCP, or FI (CFI; 217030). Esparza-Gordillo et al. (2005) suggested that complement regulatory molecules may act as a protein network, and that multiple mutations involving plasma- and membrane-associated complement regulatory proteins are necessary to impair protection of host tissues and to confer significant predisposition to aHUS.

Fremeaux-Bacchi et al. (2005) examined single-nucleotide polymorphisms (SNPs) in both the CFH and the MCP genes in 2 large cohorts of HUS. In both cohorts there was an association with HUS for both CFH and MCP alleles. Furthermore, CFH and MCP haplotypes were significantly different in HUS patients compared with controls. The results suggested that there are naturally occurring susceptibility factors in CFH and MCP for the development of atypical HUS. A characteristic feature of both MCP- and CFH-associated HUS is reduced penetrance and variable inheritance.

Caprioli et al. (2006) identified 14 mutations in the MCP gene (see, e.g., 120920.0004-120920.0006) in 20 (12.8%) of 156 patients with atypical HUS. Three patients from 1 family were compound heterozygotes, 2 patients from 1 family carried a homozygous mutation, and the others were heterozygotes. In addition, the 3 patients from 1 family also carried a mutation in the CFH gene. Most (93%) MCP mutations clustered in the 4 SCRs at the N-terminal region of MCP, indicating the importance of this region for complement regulation. The mutations resulted in either reduced protein expression or impaired C3b binding capability. Analyses of available relatives revealed a penetrance of 54%.

Associations Pending Confirmation

Feenstra et al. (2014) conducted a series of genomewide association scans comparing children with MMR-related febrile seizures, children with febrile seizures unrelated to vaccination, and controls with no history of febrile seizures. The study was restricted to individuals of Danish descent. Two loci were distinctly associated with MMR-related febrile seizures. The most associated SNP at the first locus, on chromosome 1p31.1, was rs273259 in the interferon-stimulated gene IFI44L (613975) (OR = 1.41, 95% CI 1.28-1.55, p = 5.9 x 10(-12) versus controls; OR = 1.42, 95% CI 1.27-1.59, p = 1.2 x 10(-9) versus MMR-unrelated febrile seizures). The most significant SNP at the second locus, on chromosome 1q32.2, was rs1318653, located between CD46 and CD34 (142230) (OR = 1.43, 95% CI 1.28-1.59, p = 9.6 x 10(-11) versus controls; OR = 1.48, 95% CI 1.30-1.67, p = 1.6 x 10(-9) versus MMR-unrelated febrile seizures). Feenstra et al. (2014) considered their findings implicating the innate immune system genes IFI44L and CD46 to represent a first step in understanding the biological mechanisms underlying febrile seizures as an adverse effect of MMR vaccination.


Animal Model

Marie et al. (2002) studied mice transgenic for human CD46 isoforms differing in their STP regions and in the length of their cytoplasmic domains. Mice expressing the 16-amino acid cytoplasmic tail variant, dubbed CD46-1, inhibited the T cell-mediated contact hypersensitivity reaction, whereas those expression the 23-residue cytoplasmic tail variant, termed CD46-2, increased it. CD46 stimulation or costimulation resulted in decreased cytotoxic activity and IL2 production, but increased proliferation and IL10 production, in CD46-1 transgenic mice. The effects were reversed in CD46-2 mice. Marie et al. (2002) proposed that CD46 plays a role in the regulation of the T cell-induced inflammatory reaction and in fine-tuning the cellular immune response by bridging innate and acquired immunity.


ALLELIC VARIANTS ( 6 Selected Examples):

.0001 HEMOLYTIC UREMIC SYNDROME, ATYPICAL, SUSCEPTIBILITY TO, 2

CD46, 2-BP DEL, 843AC
  
RCV000018572

In 2 sibs with diarrhea-negative atypical hemolytic uremic syndrome (AHUS2; 612922), Noris et al. (2003) identified a heterozygous mutation in the MCP gene involving deletion of nucleotides 843A and 844C. The 2-bp deletion caused a change in 3 amino acids at positions 233-235 and insertion of a premature stop codon at position 236, which resulted in loss of the C terminus of the protein. The mutation was inherited from the father, who was asymptomatic. The mutation was not found in 100 healthy controls. The proband and her brother were heterozygous for the 1160G-A polymorphism in FHR5 and were homozygous for the C variant of the polymorphism 5507C-G in CR1, associated with an expression allele (H), as described by Xiang et al. (1999).


.0002 HEMOLYTIC UREMIC SYNDROME, ATYPICAL, SUSCEPTIBILITY TO, 2

CD46, 6-BP DEL
  
RCV000018573

In 3 Belgian brothers with atypical hemolytic uremic syndrome (AHUS2; 612922) previously described by Pirson et al. (1987) and Warwicker et al. (1998), Richards et al. (2003) identified a heterozygous 6-bp deletion in the MCP gene, resulting in loss of amino acids 237 and 238. The 3 brothers were affected at the ages of 27, 31, and 35 years. The clinical features were similar in all 3. In particular, C3 levels at presentation were normal and there was no recovery of renal function. Subsequently, all 3 received a cadaver renal transplant with no recurrence of the disease. Since the original report, one of the brothers died from hepatic failure with portal hypertension of unknown etiology and one developed Waldenstrom macroglobulinemia. The mother did not carry the mutation. The father had died of pancreatic carcinoma at the age of 65 years. All 3 brothers shared a haplotype inherited from the father, an approximately 32-Mb region containing the MCP gene.


.0003 HEMOLYTIC UREMIC SYNDROME, ATYPICAL, SUSCEPTIBILITY TO, 2

CD46, SER206PRO
  
RCV000018574...

In 2 German brothers with atypical hemolytic uremic syndrome (AHUS2; 612922), Richards et al. (2003) identified a heterozygous 822T-C transition in the MCP gene, resulting in a ser206-to-pro (S206P) substitution. The mutation was inherited from the unaffected mother. The older brother presented at the age of 8 years with a short history of vomiting and findings on blood smears consistent with microangiopathic hemolytic anemia. Renal function recovered spontaneously. The younger brother presented at the age of 15 years with a 2-day history of vomiting. On admission, platelet count was greatly reduced and there was microangiopathic hemolytic anemia. Hemodialysis was initiated and the child was also treated with plasma infusions and plasma exchange. After 14 days, renal function returned and the boy made a complete recovery. Neither parent reported a history of a similar syndrome. Richards et al. (2003) also described a family from the Izmir region of Turkey in which 2 affected sibs were homozygous for the S206P mutation.


.0004 HEMOLYTIC UREMIC SYNDROME, ATYPICAL, SUSCEPTIBILITY TO, 2

CD46, IVS1AS, G-C, -1
  
RCV000018575

In 2 sibs of Sardinian origin who developed atypical hemolytic uremic syndrome (AHUS2; 612922) before age 4 years, Caprioli et al. (2006) identified a homozygous G-to-C transversion in intron 1 of the MCP gene, resulting in premature termination. Flow cytometry showed severely reduced MCP protein expression on peripheral blood cells. Their father, who developed the disorder as an adult, was heterozygous for the mutation. Four additional unaffected family members, including the mother, were also heterozygous for the mutation, indicating reduced penetrance for development of the disorder. The mutation was identified in heterozygosity in another unrelated Sardinian patient with the disorder. Haplotype analysis indicated a founder effect. The mutation was not found in 120 controls.


.0005 HEMOLYTIC UREMIC SYNDROME, ATYPICAL, SUSCEPTIBILITY TO, 2

CD46, ARG25TER
  
RCV000018576...

In 3 members of a family with atypical hemolytic uremic syndrome (AHUS2; 612922), Caprioli et al. (2006) identified compound heterozygosity for 2 mutations in the MCP gene: a 218C-T transition in exon 2 resulting in an arg25-to-ter (R25X) substitution, and C1Y (120920.0006). The R25X mutation causes loss of the entire transmembrane domain so that the protein is not expressed on the surface of peripheral blood cells. These 3 family members also carried a mutation in the CFH gene (134370). Caprioli et al. (2006) also found heterozygosity for the R25X mutation in 3 additional unrelated patients with sporadic aHUS.


.0006 HEMOLYTIC UREMIC SYNDROME, ATYPICAL, SUSCEPTIBILITY TO, 2

CD46, CYS1TYR
  
RCV000018577...

In a patient with sporadic atypical hemolytic uremic syndrome (AHUS2; 612922), Caprioli et al. (2006) identified a heterozygous 147G-A transition in exon 2 of CD46, resulting in a cys1-to-tyr (C1Y; 120920.0006) substitution. Western blot analysis indicated that the C1Y mutant protein was not expressed on the cell surface.


REFERENCES

  1. Andrews, P. W., Knowles, B. B., Parkar, M., Pym, B., Stanley, K., Goodfellow, P. N. A human cell-surface antigen defined by a monoclonal antibody and controlled by a gene on human chromosome 1. Ann. Hum. Genet. 49: 31-39, 1985. [PubMed: 2416262, related citations] [Full Text]

  2. Bora, N. S., Lublin, D. M., Kumar, B. V., Hockett, R. D., Holers, V. M., Atkinson, J. P. Structural gene for human membrane cofactor protein (MCP) of complement maps to within 100 kb of the 3-prime end of the C3b/C4b receptor gene. J. Exp. Med. 169: 597-602, 1989. [PubMed: 2521358, related citations] [Full Text]

  3. Caprioli, J., Noris, M., Brioschi, S., Pianetti, G., Castelletti, F., Bettinaglio, P., Mele, C., Bresin, E., Cassis, L., Gamba, S., Porrati, F., Bucchioni, S., Monteferrante, F., Fang, C. J., Liszewski, M. K., Kavanagh, D., Atkinson, J. P., Remuzzi, G. Genetics of HUS: the impact of MCP, CFH, and IF mutations on clinical presentation, response to treatment, and outcome. Blood 108: 1267-1279, 2006. [PubMed: 16621965, images, related citations] [Full Text]

  4. Cui, W., Hourcade, D., Post, T., Greenlund, A. C., Atkinson, J. P., Kumar, V. Characterization of the promoter region of the membrane cofactor protein (CD46) gene of the human complement system and comparison to a membrane cofactor protein-like genetic element. J. Immun. 151: 4137-4146, 1993. [PubMed: 7691939, related citations]

  5. Dorig, R. E., Marcil, A., Chopra, A., Richardson, C. D. The human CD46 molecule is a receptor for measles virus (Edmonston strain). Cell 75: 295-305, 1993. [PubMed: 8402913, related citations] [Full Text]

  6. Esparza-Gordillo, J., Goicoechea de Jorge, E., Buil, A., Berges, L. C., Lopez-Trascasa, M., Sanchez-Corral, P., Rodriguez de Cordoba, S. Predisposition to atypical hemolytic uremic syndrome involves the concurrence of different susceptibility alleles in the regulators of complement activation gene cluster in 1q32. Hum. Molec. Genet. 14: 703-712, 2005. Note: Erratum: Hum. Molec. Genet. 14: 1107 only, 2005. [PubMed: 15661753, related citations] [Full Text]

  7. Feenstra, B., Pasternak, B., Geller, F., Carstensen, L., Wang, T., Huang, F., Eitson, J. L., Hollegaard, M. V., Svanstrom, H., Vestergaard, M., Hougaard, D. M., Schoggins, J. W., Jan, L. Y., Melbye, M., Hviid, A. Common variants associated with general and MMR vaccine-related febrile seizures. Nature Genet. 46: 1274-1282, 2014. [PubMed: 25344690, related citations] [Full Text]

  8. Fremeaux-Bacchi, V., Kemp, E. J., Goodship, J. A., Dragon-Durey, M.-A., Strain, L., Loirat, C., Deng, H.-W., Goodship, T. H. J. The development of atypical haemolytic-uraemic syndrome is influenced by susceptibility factors in factor H and membrane cofactor protein: evidence from two independent cohorts. J. Med. Genet. 42: 852-856, 2005. [PubMed: 15784724, related citations] [Full Text]

  9. Gaggar, A., Shayakhmetov, D. M., Lieber, A. CD46 is a cellular receptor for group B adenoviruses. Nature Med. 9: 1408-1412, 2003. [PubMed: 14566335, related citations] [Full Text]

  10. Johansson, L., Rytkonen, A., Bergman, P., Albiger, B., Kallstrom, H., Hokfelt, T., Agerberth, B., Cattaneo, R., Jonsson, A.-B. CD46 in meningococcal disease. Science 301: 373-375, 2003. [PubMed: 12869763, related citations] [Full Text]

  11. Kallstrom, H., Gill, D. B., Albiger, B., Liszewski, M. K., Atkinson, J. P., Jonsson, A.-B. Attachment of Neisseria gonorrhoeae to the cellular pilus receptor CD46: identification of domains important for bacterial adherence. Cell. Microbiol. 3: 133-143, 2001. [PubMed: 11260136, related citations] [Full Text]

  12. Kallstrom, H., Liszewski, M. K., Atkinson, J. P., Jonsson, A.-B. Membrane cofactor protein (MCP or CD46) is a cellular pilus receptor for pathogenic Neisseria. Molec. Microbiol. 25: 639-647, 1997. [PubMed: 9379894, related citations] [Full Text]

  13. Kemper, C., Chan, A. C., Green, J. M., Brett, K. A., Murphy, K. M., Atkinson, J. P. Activation of human CD4+ cells with CD3 and CD46 induces a T-regulatory cell 1 phenotype. Nature 421: 388-392, 2003. [PubMed: 12540904, related citations] [Full Text]

  14. Lublin, D. M., Liszewski, M. K., Post, T. W., Arce, M. A., LeBeau, M. M., Rebentisch, M. B., Lemons, R. S., Seya, T., Atkinson, J. P. Molecular cloning and chromosomal localization of human complement membrane cofactor protein (MCP): evidence for inclusion in the multigene family of complement-regulatory proteins. J. Exp. Med. 168: 181-194, 1988. [PubMed: 3260937, related citations] [Full Text]

  15. Marie, J. C., Astier, A. L., Rivailler, P., Rabourdin-Combe, C., Wild, T. F., Horvat, B. Linking innate and acquired immunity: divergent role of CD46 cytoplasmic domains in T cell-induced inflammation. Nature Immun. 3: 659-666, 2002. [PubMed: 12055630, related citations] [Full Text]

  16. McIntyre, J. A., Faulk, W. P., Verhulst, S. J., Colliver, J. A. Human trophoblast-lymphocyte cross-reactive (TLX) antigens define a new alloantigen system. Science 222: 1135-1137, 1983. [PubMed: 6648525, related citations] [Full Text]

  17. Noris, M., Brioschi, S., Caprioli, J., Todeschini, M., Bresin, E., Porrati, F., Gamba, S., Remuzzi, G. Familial haemolytic uraemic syndrome and an MCP mutation. Lancet 362: 1542-1547, 2003. [PubMed: 14615110, related citations] [Full Text]

  18. Oliaro, J., Pasam, A., Waterhouse, N. J., Browne, K. A., Ludford-Menting, M. J., Trapani, J. A., Russell, S. M. Ligation of the cell surface receptor, CD46, alters T cell polarity and response to antigen presentation. Proc. Nat. Acad. Sci. 103: 18685-18690, 2006. [PubMed: 17116876, images, related citations] [Full Text]

  19. Pirson, Y., Lefebvre, C., Arnout, C., van Ypersele de Strihou, C. Hemolytic uremic syndrome in three adult siblings: a familial study and evolution. Clin. Nephrol. 28: 250-255, 1987. [PubMed: 3480783, related citations]

  20. Post, T. W., Liszewski, M. K., Adams, E. M., Tedja, I., Miller, E. A., Atkinson, J. P. Membrane cofactor protein of the complement system: alternative splicing of serine/threonine/proline-rich exons and cytoplasmic tails produces multiple isoforms that correlate with protein phenotype. J. Exp. Med. 174: 93-102, 1991. [PubMed: 1711570, related citations] [Full Text]

  21. Purcell, D. F. J., Johnstone, R. W., McKenzie, I. F. C. Identification of four different CD46 (MCP) molecules with anti-peptide antibodies. Biochem. Biophys. Res. Commun. 180: 1091-1097, 1991. [PubMed: 1953714, related citations] [Full Text]

  22. Richards, A., Kemp, E. J., Liszewski, M. K., Goodship, J. A., Lampe, A. K., Decorte, R., Muslumanoglu, M. H., Kavukcu, S., Filler, G., Pirson, Y., Wen, L. S., Atkinson, J. P., Goodship, T. H. J. Mutations in human complement regulator, membrane cofactor protein (CD46), predispose to development of familial hemolytic uremic syndrome. Proc. Nat. Acad. Sci. 100: 12966-12971, 2003. [PubMed: 14566051, images, related citations] [Full Text]

  23. Santoro, F., Kennedy, P. E., Locatelli, G., Malnati, M. S., Berger, E. A., Lusso, P. CD46 is a cellular receptor for human herpesvirus 6. Cell 99: 817-827, 1999. [PubMed: 10619434, related citations] [Full Text]

  24. Sood, R., Zehnder, J. L., Druzin, M. L., Brown, P. O. Gene expression patterns in human placenta. Proc. Nat. Acad. Sci. 103: 5478-5483, 2006. [PubMed: 16567644, images, related citations] [Full Text]

  25. Tatsuo, H., Ono, N., Tanaka, K., Yanagi, Y. SLAM (CDw150) is a cellular receptor for measles virus. Nature 406: 893-897, 2000. [PubMed: 10972291, related citations] [Full Text]

  26. Warwicker, P., Goodship, T. H. J., Donne, R. L., Pirson, Y., Nicholls, A., Ward, R. M., Turnpenny, P., Goodship, J. A. Genetic studies into inherited and sporadic hemolytic uremic syndrome. Kidney Int. 53: 836-844, 1998. [PubMed: 9551389, related citations] [Full Text]

  27. Xiang, L., Rundles, J. R., Hamilton, D. R., Wilson, J. G. Quantitative alleles of CRI: coding sequence analysis and comparison of haplotypes in two ethnic groups. J. Immun. 163: 4939-4945, 1999. [PubMed: 10528197, related citations]


Ada Hamosh - updated : 03/19/2018
Paul J. Converse - updated : 4/20/2011
Cassandra L. Kniffin - updated : 7/27/2009
George E. Tiller - updated : 2/5/2008
Paul J. Converse - updated : 5/2/2007
Anne M. Stumpf - updated : 8/8/2006
Ada Hamosh - updated : 8/8/2006
Victor A. McKusick - updated : 6/22/2006
Victor A. McKusick - updated : 7/16/2004
Victor A. McKusick - updated : 2/9/2004
Paul J. Converse - updated : 10/30/2003
Ada Hamosh - updated : 8/5/2003
Paul J. Converse - updated : 1/22/2003
Paul J. Converse - updated : 6/11/2002
Paul J. Converse - updated : 4/29/2002
Paul J. Converse - updated : 4/15/2002
Paul J. Converse - updated : 8/23/2000
Stylianos E. Antonarakis - updated : 1/19/2000
Creation Date:
Victor A. McKusick : 6/29/1988
carol : 03/20/2018
alopez : 03/19/2018
carol : 09/18/2013
ckniffin : 5/9/2012
carol : 4/30/2012
carol : 11/28/2011
carol : 4/20/2011
carol : 2/28/2011
mgross : 8/18/2009
carol : 7/30/2009
ckniffin : 7/27/2009
wwang : 2/13/2008
terry : 2/5/2008
mgross : 5/2/2007
ckniffin : 10/5/2006
alopez : 8/8/2006
alopez : 8/8/2006
joanna : 7/18/2006
alopez : 6/28/2006
terry : 6/22/2006
wwang : 8/2/2005
terry : 11/3/2004
tkritzer : 7/21/2004
terry : 7/16/2004
carol : 4/28/2004
tkritzer : 4/20/2004
mgross : 3/17/2004
cwells : 2/17/2004
terry : 2/9/2004
alopez : 11/7/2003
mgross : 10/30/2003
tkritzer : 10/7/2003
alopez : 8/7/2003
terry : 8/5/2003
mgross : 1/23/2003
mgross : 1/22/2003
alopez : 7/25/2002
mgross : 6/11/2002
mgross : 4/29/2002
mgross : 4/29/2002
mgross : 4/15/2002
mgross : 8/23/2000
mgross : 1/19/2000
alopez : 4/1/1999
carol : 11/2/1993
supermim : 3/16/1992
carol : 1/21/1992
supermim : 3/20/1990
ddp : 10/26/1989
root : 3/13/1989

* 120920

CD46 ANTIGEN; CD46


Alternative titles; symbols

TROPHOBLAST-LYMPHOCYTE CROSS-REACTIVE ANTIGEN; TLX
MEMBRANE COFACTOR PROTEIN; MCP
COMPLEMENT MEMBRANE COFACTOR PROTEIN
ANTIGEN DEFINED BY MONOCLONAL ANTIBODY TRA-2-10; MIC10


HGNC Approved Gene Symbol: CD46

Cytogenetic location: 1q32.2     Genomic coordinates (GRCh38): 1:207,752,038-207,795,516 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
1q32.2 {Hemolytic uremic syndrome, atypical, susceptibility to, 2} 612922 Autosomal dominant; Autosomal recessive 3

TEXT

Description

MCP, a C3B/C4B-binding molecule of the complement system with cofactor activity for the I-dependent cleavage of C3B and C4B, is widely distributed in white blood cells, platelets, epithelial cells, and fibroblasts (Lublin et al., 1988).


Cloning and Expression

Lublin et al. (1988) purified MCP from a human T-cell line and determined the sequence of the N-terminal 24 amino acids. An oligonucleotide probe was used to identify a clone from a human monocyte cDNA library. The deduced full-length MCP consists of a 34-amino acid signal peptide and a 350-amino acid mature protein. The protein has, beginning at the N terminus, 4 cysteine-rich repeating units (short consensus repeats, or SCRs) of about 60 amino acids each that match the consensus sequence found in a multigene family of complement regulatory proteins: CR1 (120620), CR2 (120650), C4BP (120830), CFH (134370), and DAF (125240). Immediately C-terminal of the SCRs is a serine/threonine/proline (STP)-rich region, a likely area for extensive O-glycosylation. MCP also has a transmembrane domain, a basic amino acid anchor, and a cytoplasmic tail.

Purcell et al. (1991) and Post et al. (1991) identified 4 and 6 isoforms of MCP, respectively. Post et al. (1991) demonstrated that the 6 isoforms vary in having 1 of 2 cytoplasmic tails and by having either all 3 STP regions (termed A, B, and C) or only STP-BC or STP-C. They showed that the STP-C isoforms are expressed as 45- to 55-kD proteins, the STP-BC isoforms are expressed as 55- to 65-kD proteins, and the STP-ABC isoforms are expressed as 65- to 75-kD proteins. The 65- to 75-kD variants were not expressed on peripheral blood cells or cell lines. Post et al. (1991) concluded that the presence of the B region of the STP area, which is richer in O-linked sugars, determines the expression of the 2 broad protein species. They also noted that up to 4 different forms of MCP are expressed on a single cell.


Gene Function

McIntyre et al. (1983) found that antisera to human syncytiotrophoblast microvillus cell surface membranes from different placentas are cytotoxic for lymphocytes from some persons but not others. Study of 10 antisera on lymphocytes from 30 donors suggested the presence of 3 distinct TLX groupings. McIntyre et al. (1983) proposed that TLX alloantigens are central in establishing maternal recognition and protection of the blastocyst, and that lack of recognition results in implantation failure and spontaneous abortion.

Dorig et al. (1993) used a genetic approach to identify the receptor for measles virus (MV). They tested human/rodent somatic cell hybrids for their ability to bind the Edmonston strain of MV and found that only cells containing human chromosome 1 were capable of binding virus. Rodent cells could not bind MV. A study of lymphocyte markers suggested that CD46 is the MV receptor. Dorig et al. (1993) showed that hamster cell lines expressing human CD46 could bind MV. Furthermore, infected CD46+ cells produced syncytia and viral proteins. Finally, polyclonal antisera against CD46 inhibited MV binding and infection. Dorig et al. (1993) concluded that CD46 is receptor for the Edmonston strain of MV. However, Tatsuo et al. (2000) noted that although the Edmonston strain of MV and the vaccine strains derived from it use CD46 as a cellular receptor, most clinical isolates do not. They showed that SLAM (603492), but not CD46, could act as a cellular receptor for clinical strains of MV.

Human herpesvirus-6 (HHV-6) is the etiologic agent of exanthema subitum, causes opportunistic infections in immunocompromised patients, and has been implicated in multiple sclerosis and in the progression of AIDS. Santoro et al. (1999) showed that the 2 major HHV-6 subgroups (A and B) use human CD46 as a cellular receptor. Downregulation of surface CD46 was documented during the course of HHV-6 infection. Both acute infection and cell fusion mediated by HHV-6 were specifically inhibited by a monoclonal antibody to CD46; fusion was also blocked by soluble CD46. Nonhuman cells that were resistant to HHV-6 fusion and entry became susceptible upon expression of recombinant human CD46.

CD46 acts as a cellular receptor for type IV pili of pathogenic Neisseria. The binding of piliated bacteria to host cells can be inhibited by antibody to CD46 or the recombinant molecule. Kallstrom et al. (2001) showed that the BC1 phenotype of the STP domain as well as the complement control protein repeat 3 (CCP3) of CD46 are important for efficient adherence of N. gonorrhoeae to host cells.

Kemper et al. (2003) examined the requirements for activation of T-regulatory-1 (Tr1) cells, which are defined as CD4 (186940)-positive T lymphocytes that secrete IL10 (124092) and suppress T-helper cells. Stimulation of purified CD4-positive T cells with monoclonal antibodies to CD3 (see 186740) and CD46 in the presence of IL2 (147680) or anti-CD28 (186760) induced the secretion of large amounts of IL10 and sustained proliferation, as measured by flow cytometric analysis for expression of PCNA (176740). CD45RA-positive/CD45RO-negative (naive) T cells and CD45RA-positive/CD45RO-positive (high-responding) T cells produced IL10 in response to these agonists, while CD45RA-negative/CD45RO-positive (memory) T cells did not. After primary anti-CD3/anti-CD46 activation, however, both naive and high-responding CD4-positive T cells acquired an IL10-producing memory phenotype (CD45RA-negative/CD45RO-positive). Stimulation of CD4-positive T cells with anti-CD3/anti-CD28 without anti-CD46 failed to induce IL10 production and caused the production of large amounts of IL2. Stimulation with anti-CD3/anti-CD28 in the presence of complement factor C3b (120700) dimers resulted in IL10 secretion comparable to that of anti-CD3/anti-CD28/anti-CD46-activated T cells. Supernatants of the anti-CD3/anti-CD46-activated T cells induced IL10-mediated suppression of proliferation by bystander T cells. Kemper et al. (2003) concluded that CD46 has a role in human T-cell regulation and that these findings establish a link between the complement system and adaptive immunity. They proposed that Tr1 cells are essential for maintaining peripheral tolerance and preventing autoimmunity, as well as for responses to many pathogens.

Kallstrom et al. (1997) identified CD46 as a human cell surface receptor for piliated pathogenic Neisseria. Johansson et al. (2003) generated transgenic mice expressing human CD46 and found they were susceptible to meningococcal disease because bacteria crossed the blood-brain barrier in these mice. Development of disease was more efficient with piliated bacteria after intranasal but not intraperitoneal challenge of Cd46 transgenic mice, suggesting that human CD46 facilitates pilus-dependent interactions at the epithelial mucosa.

Gaggar et al. (2003) showed that, unlike most adenoviruses, group B adenoviruses use CD46 rather than CAR (CXADR; 602621) to infect cells. Mass spectrometric, immunoblot, and fluorescence microscopy analyses determined that the group B fiber knob domain interacts with CD46. The authors found that, with the exception of Ad3, viruses of the B1 respiratory and B2 kidney and urinary tract subgroups both use the CD46 receptor. Expression of human CD46 rendered nonhuman cells susceptible to group B adenovirus infection in vitro and in vivo, and infection could be blocked by CD46 siRNA or soluble CD46 protein. Gaggar et al. (2003) concluded that CD46 is an essential receptor for group B adenoviruses and that this knowledge may facilitate the development of novel methods for adenovirus-mediated gene transfer.

The placenta is an immunologically privileged site. Using DNA microarrays to compare gene expression patterns, Sood et al. (2006) found that 3 regulators of complement, CD55 (125240), CD59 (107271), and MCP, are expressed at higher levels in normal placental villus sections compared with other normal human tissues. Within the placenta, CD55 and CD59 are expressed at greatest levels in amnion, followed by chorion and villus sections, whereas MCP is expressed at higher levels only in villus sections. These inhibitors of complement are expressed on syncytiotrophoblasts, the specialized placental cells lining the villi that are in direct contact with maternal blood. The amnion compared with chorion is remarkably nonimmunogenic, and the immune properties of the amnion are intriguing because it is not in direct contact with maternal cells. Sood et al. (2006) suggested that the amnion may secrete the complement inhibitors themselves or in the form of protected exosomes into the amniotic fluid or the neighboring maternofetal junction.

Using confocal microscopy, Oliaro et al. (2006) demonstrated that treatment of T or natural killer (NK) cytotoxic lymphocytes with anti-CD46 beads, but not anti-transferrin receptor (TFRC; 190010) beads, resulted in recruitment of the microtubule-organizing center (MTOC) and perforin (PRF1; 170280) to the site of CD46 ligation, indicating cell polarization. Ligation with soluble anti-CD46 altered T-cell polarization, inhibiting IFNG (147570) production. CD46 ligation also prevented normal polarization of T cells towards antigen-presenting cells (APCs) and prevented immune synapse formation, but it did not affect interaction of NK cells with target cells. Cells expressing measles hemagglutinin ligated CD46 on T cells and recruited the MTOC and CD3 to the site. These T cells only produced IFNG with the addition of anti-CD3/CD28. Oliaro et al. (2006) concluded that external signals can alter lymphocyte polarization toward APC or target cells and inhibit lymphocyte function.


Gene Structure

By genomic sequence analysis, Post et al. (1991) determined that the MCP gene contains 14 exons and spans 43 kb. Exons 7, 8, and 9 encode the A, B, and C regions of the STP area, respectively. Exon 13 encodes a 16-residue cytoplasmic tail, whereas use of exon 14 results in a 23-amino acid tail.

Cui et al. (1993) analyzed the promoter region of the CD46 gene. They detected a GC-rich region as well as a CAAT box in reverse orientation surrounded by 4 putative SP1-binding sites, but no TATA element, suggesting that CD46 is a housekeeping gene. Reporter assays determined that promoter activity is confined to the GC-rich region.


Mapping

Andrews et al. (1985) studied an antigen expressed by most human cells, but not erythrocytes, and defined by monoclonal antibody TRA-2-10. The antigen was expressed on the surface of human-mouse somatic cell hybrids; segregation analysis showed that the antigen is determined by a gene (MCP1) on human chromosome 1.

Lublin et al. (1988) localized the MCP gene to 1q31-q41 by Southern analysis of human-rodent somatic cell hybrid DNA and by in situ hybridization. This was the sixth member of this multigene family that had been assigned to this region of the genome. Bora et al. (1989) demonstrated that the MCP gene is on the same 1,250-kb NotI fragment that contains CR1, CR2, DAF, and C4BP and maps within 100 kb of the 3-prime end of the CR1 gene. The order of the genes appears to be that just indicated, with MCP preceding the other 4 genes.


Molecular Genetics

Atypical Hemolytic Uremic Syndrome, Susceptibility to, 2

Noris et al. (2003) identified a heterozygous mutation (120920.0001) in the MCP gene in 2 patients with a family history of atypical hemolytic uremic syndrome (AHUS2; 612922). The mutation caused a change in 3 amino acids at position 233-235 and insertion of a premature stop codon, which resulted in loss of the transmembrane domain of the protein and severely reduced cell-surface expression of MCP.

Like factor H (CFH; 134370), MCP inhibits complement activation by regulating C3b deposition on targets. Richards et al. (2003) hypothesized that MCP mutations could predispose to aHUS, and they sequenced MCP coding exons in affected members from 30 affected families. They identified mutations in the MCP gene in affected members from 3 of these families: a heterozygous 6-bp deletion in one family (120920.0002) and a ser206-to-pro mutation (S206P; 120920.0003), which was heterozygous in one family and homozygous in another. An individual with the 6-bp deletion had reduced MCP levels and approximately 50% C3b binding compared with normal controls. Individuals with the S206P mutation expressed normal quantities of protein, but demonstrated approximately 50% reduction in C3b binding in heterozygotes and complete lack of C3b binding in homozygotes. Studies in transfectants showed that the deletion mutant was retained intracellularly. S206P protein was expressed on the cell surface but had a reduced ability to prevent complement activation, consistent with its reduced C3b binding and cofactor activity.

Esparza-Gordillo et al. (2005) identified a specific SNP haplotype in the MCP gene, which was overrepresented in aHUS patients and strongly associated with the severity of the disease. Linkage disequilibrium analysis suggested that the haplotype included the CR1 (120620), DAF (CD55; 125240), and C4BPA (120830) genes. Two SNPs in the haplotype influenced the transcription activity of the MCP promoter in transient transfection experiments. The SNP haplotype block was particularly frequent among patients who carried mutations in HF1 (CFH; 134370), MCP, or FI (CFI; 217030). Esparza-Gordillo et al. (2005) suggested that complement regulatory molecules may act as a protein network, and that multiple mutations involving plasma- and membrane-associated complement regulatory proteins are necessary to impair protection of host tissues and to confer significant predisposition to aHUS.

Fremeaux-Bacchi et al. (2005) examined single-nucleotide polymorphisms (SNPs) in both the CFH and the MCP genes in 2 large cohorts of HUS. In both cohorts there was an association with HUS for both CFH and MCP alleles. Furthermore, CFH and MCP haplotypes were significantly different in HUS patients compared with controls. The results suggested that there are naturally occurring susceptibility factors in CFH and MCP for the development of atypical HUS. A characteristic feature of both MCP- and CFH-associated HUS is reduced penetrance and variable inheritance.

Caprioli et al. (2006) identified 14 mutations in the MCP gene (see, e.g., 120920.0004-120920.0006) in 20 (12.8%) of 156 patients with atypical HUS. Three patients from 1 family were compound heterozygotes, 2 patients from 1 family carried a homozygous mutation, and the others were heterozygotes. In addition, the 3 patients from 1 family also carried a mutation in the CFH gene. Most (93%) MCP mutations clustered in the 4 SCRs at the N-terminal region of MCP, indicating the importance of this region for complement regulation. The mutations resulted in either reduced protein expression or impaired C3b binding capability. Analyses of available relatives revealed a penetrance of 54%.

Associations Pending Confirmation

Feenstra et al. (2014) conducted a series of genomewide association scans comparing children with MMR-related febrile seizures, children with febrile seizures unrelated to vaccination, and controls with no history of febrile seizures. The study was restricted to individuals of Danish descent. Two loci were distinctly associated with MMR-related febrile seizures. The most associated SNP at the first locus, on chromosome 1p31.1, was rs273259 in the interferon-stimulated gene IFI44L (613975) (OR = 1.41, 95% CI 1.28-1.55, p = 5.9 x 10(-12) versus controls; OR = 1.42, 95% CI 1.27-1.59, p = 1.2 x 10(-9) versus MMR-unrelated febrile seizures). The most significant SNP at the second locus, on chromosome 1q32.2, was rs1318653, located between CD46 and CD34 (142230) (OR = 1.43, 95% CI 1.28-1.59, p = 9.6 x 10(-11) versus controls; OR = 1.48, 95% CI 1.30-1.67, p = 1.6 x 10(-9) versus MMR-unrelated febrile seizures). Feenstra et al. (2014) considered their findings implicating the innate immune system genes IFI44L and CD46 to represent a first step in understanding the biological mechanisms underlying febrile seizures as an adverse effect of MMR vaccination.


Animal Model

Marie et al. (2002) studied mice transgenic for human CD46 isoforms differing in their STP regions and in the length of their cytoplasmic domains. Mice expressing the 16-amino acid cytoplasmic tail variant, dubbed CD46-1, inhibited the T cell-mediated contact hypersensitivity reaction, whereas those expression the 23-residue cytoplasmic tail variant, termed CD46-2, increased it. CD46 stimulation or costimulation resulted in decreased cytotoxic activity and IL2 production, but increased proliferation and IL10 production, in CD46-1 transgenic mice. The effects were reversed in CD46-2 mice. Marie et al. (2002) proposed that CD46 plays a role in the regulation of the T cell-induced inflammatory reaction and in fine-tuning the cellular immune response by bridging innate and acquired immunity.


ALLELIC VARIANTS 6 Selected Examples):

.0001   HEMOLYTIC UREMIC SYNDROME, ATYPICAL, SUSCEPTIBILITY TO, 2

CD46, 2-BP DEL, 843AC
SNP: rs1558056827, ClinVar: RCV000018572

In 2 sibs with diarrhea-negative atypical hemolytic uremic syndrome (AHUS2; 612922), Noris et al. (2003) identified a heterozygous mutation in the MCP gene involving deletion of nucleotides 843A and 844C. The 2-bp deletion caused a change in 3 amino acids at positions 233-235 and insertion of a premature stop codon at position 236, which resulted in loss of the C terminus of the protein. The mutation was inherited from the father, who was asymptomatic. The mutation was not found in 100 healthy controls. The proband and her brother were heterozygous for the 1160G-A polymorphism in FHR5 and were homozygous for the C variant of the polymorphism 5507C-G in CR1, associated with an expression allele (H), as described by Xiang et al. (1999).


.0002   HEMOLYTIC UREMIC SYNDROME, ATYPICAL, SUSCEPTIBILITY TO, 2

CD46, 6-BP DEL
SNP: rs1255421232, gnomAD: rs1255421232, ClinVar: RCV000018573

In 3 Belgian brothers with atypical hemolytic uremic syndrome (AHUS2; 612922) previously described by Pirson et al. (1987) and Warwicker et al. (1998), Richards et al. (2003) identified a heterozygous 6-bp deletion in the MCP gene, resulting in loss of amino acids 237 and 238. The 3 brothers were affected at the ages of 27, 31, and 35 years. The clinical features were similar in all 3. In particular, C3 levels at presentation were normal and there was no recovery of renal function. Subsequently, all 3 received a cadaver renal transplant with no recurrence of the disease. Since the original report, one of the brothers died from hepatic failure with portal hypertension of unknown etiology and one developed Waldenstrom macroglobulinemia. The mother did not carry the mutation. The father had died of pancreatic carcinoma at the age of 65 years. All 3 brothers shared a haplotype inherited from the father, an approximately 32-Mb region containing the MCP gene.


.0003   HEMOLYTIC UREMIC SYNDROME, ATYPICAL, SUSCEPTIBILITY TO, 2

CD46, SER206PRO
SNP: rs121909589, gnomAD: rs121909589, ClinVar: RCV000018574, RCV002514110

In 2 German brothers with atypical hemolytic uremic syndrome (AHUS2; 612922), Richards et al. (2003) identified a heterozygous 822T-C transition in the MCP gene, resulting in a ser206-to-pro (S206P) substitution. The mutation was inherited from the unaffected mother. The older brother presented at the age of 8 years with a short history of vomiting and findings on blood smears consistent with microangiopathic hemolytic anemia. Renal function recovered spontaneously. The younger brother presented at the age of 15 years with a 2-day history of vomiting. On admission, platelet count was greatly reduced and there was microangiopathic hemolytic anemia. Hemodialysis was initiated and the child was also treated with plasma infusions and plasma exchange. After 14 days, renal function returned and the boy made a complete recovery. Neither parent reported a history of a similar syndrome. Richards et al. (2003) also described a family from the Izmir region of Turkey in which 2 affected sibs were homozygous for the S206P mutation.


.0004   HEMOLYTIC UREMIC SYNDROME, ATYPICAL, SUSCEPTIBILITY TO, 2

CD46, IVS1AS, G-C, -1
SNP: rs1441937053, ClinVar: RCV000018575

In 2 sibs of Sardinian origin who developed atypical hemolytic uremic syndrome (AHUS2; 612922) before age 4 years, Caprioli et al. (2006) identified a homozygous G-to-C transversion in intron 1 of the MCP gene, resulting in premature termination. Flow cytometry showed severely reduced MCP protein expression on peripheral blood cells. Their father, who developed the disorder as an adult, was heterozygous for the mutation. Four additional unaffected family members, including the mother, were also heterozygous for the mutation, indicating reduced penetrance for development of the disorder. The mutation was identified in heterozygosity in another unrelated Sardinian patient with the disorder. Haplotype analysis indicated a founder effect. The mutation was not found in 120 controls.


.0005   HEMOLYTIC UREMIC SYNDROME, ATYPICAL, SUSCEPTIBILITY TO, 2

CD46, ARG25TER
SNP: rs121909590, gnomAD: rs121909590, ClinVar: RCV000018576, RCV001508038

In 3 members of a family with atypical hemolytic uremic syndrome (AHUS2; 612922), Caprioli et al. (2006) identified compound heterozygosity for 2 mutations in the MCP gene: a 218C-T transition in exon 2 resulting in an arg25-to-ter (R25X) substitution, and C1Y (120920.0006). The R25X mutation causes loss of the entire transmembrane domain so that the protein is not expressed on the surface of peripheral blood cells. These 3 family members also carried a mutation in the CFH gene (134370). Caprioli et al. (2006) also found heterozygosity for the R25X mutation in 3 additional unrelated patients with sporadic aHUS.


.0006   HEMOLYTIC UREMIC SYNDROME, ATYPICAL, SUSCEPTIBILITY TO, 2

CD46, CYS1TYR
SNP: rs121909591, gnomAD: rs121909591, ClinVar: RCV000018577, RCV002513104

In a patient with sporadic atypical hemolytic uremic syndrome (AHUS2; 612922), Caprioli et al. (2006) identified a heterozygous 147G-A transition in exon 2 of CD46, resulting in a cys1-to-tyr (C1Y; 120920.0006) substitution. Western blot analysis indicated that the C1Y mutant protein was not expressed on the cell surface.


REFERENCES

  1. Andrews, P. W., Knowles, B. B., Parkar, M., Pym, B., Stanley, K., Goodfellow, P. N. A human cell-surface antigen defined by a monoclonal antibody and controlled by a gene on human chromosome 1. Ann. Hum. Genet. 49: 31-39, 1985. [PubMed: 2416262] [Full Text: https://doi.org/10.1111/j.1469-1809.1985.tb01673.x]

  2. Bora, N. S., Lublin, D. M., Kumar, B. V., Hockett, R. D., Holers, V. M., Atkinson, J. P. Structural gene for human membrane cofactor protein (MCP) of complement maps to within 100 kb of the 3-prime end of the C3b/C4b receptor gene. J. Exp. Med. 169: 597-602, 1989. [PubMed: 2521358] [Full Text: https://doi.org/10.1084/jem.169.2.597]

  3. Caprioli, J., Noris, M., Brioschi, S., Pianetti, G., Castelletti, F., Bettinaglio, P., Mele, C., Bresin, E., Cassis, L., Gamba, S., Porrati, F., Bucchioni, S., Monteferrante, F., Fang, C. J., Liszewski, M. K., Kavanagh, D., Atkinson, J. P., Remuzzi, G. Genetics of HUS: the impact of MCP, CFH, and IF mutations on clinical presentation, response to treatment, and outcome. Blood 108: 1267-1279, 2006. [PubMed: 16621965] [Full Text: https://doi.org/10.1182/blood-2005-10-007252]

  4. Cui, W., Hourcade, D., Post, T., Greenlund, A. C., Atkinson, J. P., Kumar, V. Characterization of the promoter region of the membrane cofactor protein (CD46) gene of the human complement system and comparison to a membrane cofactor protein-like genetic element. J. Immun. 151: 4137-4146, 1993. [PubMed: 7691939]

  5. Dorig, R. E., Marcil, A., Chopra, A., Richardson, C. D. The human CD46 molecule is a receptor for measles virus (Edmonston strain). Cell 75: 295-305, 1993. [PubMed: 8402913] [Full Text: https://doi.org/10.1016/0092-8674(93)80071-l]

  6. Esparza-Gordillo, J., Goicoechea de Jorge, E., Buil, A., Berges, L. C., Lopez-Trascasa, M., Sanchez-Corral, P., Rodriguez de Cordoba, S. Predisposition to atypical hemolytic uremic syndrome involves the concurrence of different susceptibility alleles in the regulators of complement activation gene cluster in 1q32. Hum. Molec. Genet. 14: 703-712, 2005. Note: Erratum: Hum. Molec. Genet. 14: 1107 only, 2005. [PubMed: 15661753] [Full Text: https://doi.org/10.1093/hmg/ddi066]

  7. Feenstra, B., Pasternak, B., Geller, F., Carstensen, L., Wang, T., Huang, F., Eitson, J. L., Hollegaard, M. V., Svanstrom, H., Vestergaard, M., Hougaard, D. M., Schoggins, J. W., Jan, L. Y., Melbye, M., Hviid, A. Common variants associated with general and MMR vaccine-related febrile seizures. Nature Genet. 46: 1274-1282, 2014. [PubMed: 25344690] [Full Text: https://doi.org/10.1038/ng.3129]

  8. Fremeaux-Bacchi, V., Kemp, E. J., Goodship, J. A., Dragon-Durey, M.-A., Strain, L., Loirat, C., Deng, H.-W., Goodship, T. H. J. The development of atypical haemolytic-uraemic syndrome is influenced by susceptibility factors in factor H and membrane cofactor protein: evidence from two independent cohorts. J. Med. Genet. 42: 852-856, 2005. [PubMed: 15784724] [Full Text: https://doi.org/10.1136/jmg.2005.030783]

  9. Gaggar, A., Shayakhmetov, D. M., Lieber, A. CD46 is a cellular receptor for group B adenoviruses. Nature Med. 9: 1408-1412, 2003. [PubMed: 14566335] [Full Text: https://doi.org/10.1038/nm952]

  10. Johansson, L., Rytkonen, A., Bergman, P., Albiger, B., Kallstrom, H., Hokfelt, T., Agerberth, B., Cattaneo, R., Jonsson, A.-B. CD46 in meningococcal disease. Science 301: 373-375, 2003. [PubMed: 12869763] [Full Text: https://doi.org/10.1126/science.1086476]

  11. Kallstrom, H., Gill, D. B., Albiger, B., Liszewski, M. K., Atkinson, J. P., Jonsson, A.-B. Attachment of Neisseria gonorrhoeae to the cellular pilus receptor CD46: identification of domains important for bacterial adherence. Cell. Microbiol. 3: 133-143, 2001. [PubMed: 11260136] [Full Text: https://doi.org/10.1046/j.1462-5822.2001.00095.x]

  12. Kallstrom, H., Liszewski, M. K., Atkinson, J. P., Jonsson, A.-B. Membrane cofactor protein (MCP or CD46) is a cellular pilus receptor for pathogenic Neisseria. Molec. Microbiol. 25: 639-647, 1997. [PubMed: 9379894] [Full Text: https://doi.org/10.1046/j.1365-2958.1997.4841857.x]

  13. Kemper, C., Chan, A. C., Green, J. M., Brett, K. A., Murphy, K. M., Atkinson, J. P. Activation of human CD4+ cells with CD3 and CD46 induces a T-regulatory cell 1 phenotype. Nature 421: 388-392, 2003. [PubMed: 12540904] [Full Text: https://doi.org/10.1038/nature01315]

  14. Lublin, D. M., Liszewski, M. K., Post, T. W., Arce, M. A., LeBeau, M. M., Rebentisch, M. B., Lemons, R. S., Seya, T., Atkinson, J. P. Molecular cloning and chromosomal localization of human complement membrane cofactor protein (MCP): evidence for inclusion in the multigene family of complement-regulatory proteins. J. Exp. Med. 168: 181-194, 1988. [PubMed: 3260937] [Full Text: https://doi.org/10.1084/jem.168.1.181]

  15. Marie, J. C., Astier, A. L., Rivailler, P., Rabourdin-Combe, C., Wild, T. F., Horvat, B. Linking innate and acquired immunity: divergent role of CD46 cytoplasmic domains in T cell-induced inflammation. Nature Immun. 3: 659-666, 2002. [PubMed: 12055630] [Full Text: https://doi.org/10.1038/ni810]

  16. McIntyre, J. A., Faulk, W. P., Verhulst, S. J., Colliver, J. A. Human trophoblast-lymphocyte cross-reactive (TLX) antigens define a new alloantigen system. Science 222: 1135-1137, 1983. [PubMed: 6648525] [Full Text: https://doi.org/10.1126/science.6648525]

  17. Noris, M., Brioschi, S., Caprioli, J., Todeschini, M., Bresin, E., Porrati, F., Gamba, S., Remuzzi, G. Familial haemolytic uraemic syndrome and an MCP mutation. Lancet 362: 1542-1547, 2003. [PubMed: 14615110] [Full Text: https://doi.org/10.1016/S0140-6736(03)14742-3]

  18. Oliaro, J., Pasam, A., Waterhouse, N. J., Browne, K. A., Ludford-Menting, M. J., Trapani, J. A., Russell, S. M. Ligation of the cell surface receptor, CD46, alters T cell polarity and response to antigen presentation. Proc. Nat. Acad. Sci. 103: 18685-18690, 2006. [PubMed: 17116876] [Full Text: https://doi.org/10.1073/pnas.0602458103]

  19. Pirson, Y., Lefebvre, C., Arnout, C., van Ypersele de Strihou, C. Hemolytic uremic syndrome in three adult siblings: a familial study and evolution. Clin. Nephrol. 28: 250-255, 1987. [PubMed: 3480783]

  20. Post, T. W., Liszewski, M. K., Adams, E. M., Tedja, I., Miller, E. A., Atkinson, J. P. Membrane cofactor protein of the complement system: alternative splicing of serine/threonine/proline-rich exons and cytoplasmic tails produces multiple isoforms that correlate with protein phenotype. J. Exp. Med. 174: 93-102, 1991. [PubMed: 1711570] [Full Text: https://doi.org/10.1084/jem.174.1.93]

  21. Purcell, D. F. J., Johnstone, R. W., McKenzie, I. F. C. Identification of four different CD46 (MCP) molecules with anti-peptide antibodies. Biochem. Biophys. Res. Commun. 180: 1091-1097, 1991. [PubMed: 1953714] [Full Text: https://doi.org/10.1016/s0006-291x(05)81178-7]

  22. Richards, A., Kemp, E. J., Liszewski, M. K., Goodship, J. A., Lampe, A. K., Decorte, R., Muslumanoglu, M. H., Kavukcu, S., Filler, G., Pirson, Y., Wen, L. S., Atkinson, J. P., Goodship, T. H. J. Mutations in human complement regulator, membrane cofactor protein (CD46), predispose to development of familial hemolytic uremic syndrome. Proc. Nat. Acad. Sci. 100: 12966-12971, 2003. [PubMed: 14566051] [Full Text: https://doi.org/10.1073/pnas.2135497100]

  23. Santoro, F., Kennedy, P. E., Locatelli, G., Malnati, M. S., Berger, E. A., Lusso, P. CD46 is a cellular receptor for human herpesvirus 6. Cell 99: 817-827, 1999. [PubMed: 10619434] [Full Text: https://doi.org/10.1016/s0092-8674(00)81678-5]

  24. Sood, R., Zehnder, J. L., Druzin, M. L., Brown, P. O. Gene expression patterns in human placenta. Proc. Nat. Acad. Sci. 103: 5478-5483, 2006. [PubMed: 16567644] [Full Text: https://doi.org/10.1073/pnas.0508035103]

  25. Tatsuo, H., Ono, N., Tanaka, K., Yanagi, Y. SLAM (CDw150) is a cellular receptor for measles virus. Nature 406: 893-897, 2000. [PubMed: 10972291] [Full Text: https://doi.org/10.1038/35022579]

  26. Warwicker, P., Goodship, T. H. J., Donne, R. L., Pirson, Y., Nicholls, A., Ward, R. M., Turnpenny, P., Goodship, J. A. Genetic studies into inherited and sporadic hemolytic uremic syndrome. Kidney Int. 53: 836-844, 1998. [PubMed: 9551389] [Full Text: https://doi.org/10.1111/j.1523-1755.1998.00824.x]

  27. Xiang, L., Rundles, J. R., Hamilton, D. R., Wilson, J. G. Quantitative alleles of CRI: coding sequence analysis and comparison of haplotypes in two ethnic groups. J. Immun. 163: 4939-4945, 1999. [PubMed: 10528197]


Contributors:
Ada Hamosh - updated : 03/19/2018
Paul J. Converse - updated : 4/20/2011
Cassandra L. Kniffin - updated : 7/27/2009
George E. Tiller - updated : 2/5/2008
Paul J. Converse - updated : 5/2/2007
Anne M. Stumpf - updated : 8/8/2006
Ada Hamosh - updated : 8/8/2006
Victor A. McKusick - updated : 6/22/2006
Victor A. McKusick - updated : 7/16/2004
Victor A. McKusick - updated : 2/9/2004
Paul J. Converse - updated : 10/30/2003
Ada Hamosh - updated : 8/5/2003
Paul J. Converse - updated : 1/22/2003
Paul J. Converse - updated : 6/11/2002
Paul J. Converse - updated : 4/29/2002
Paul J. Converse - updated : 4/15/2002
Paul J. Converse - updated : 8/23/2000
Stylianos E. Antonarakis - updated : 1/19/2000

Creation Date:
Victor A. McKusick : 6/29/1988

Edit History:
carol : 03/20/2018
alopez : 03/19/2018
carol : 09/18/2013
ckniffin : 5/9/2012
carol : 4/30/2012
carol : 11/28/2011
carol : 4/20/2011
carol : 2/28/2011
mgross : 8/18/2009
carol : 7/30/2009
ckniffin : 7/27/2009
wwang : 2/13/2008
terry : 2/5/2008
mgross : 5/2/2007
ckniffin : 10/5/2006
alopez : 8/8/2006
alopez : 8/8/2006
joanna : 7/18/2006
alopez : 6/28/2006
terry : 6/22/2006
wwang : 8/2/2005
terry : 11/3/2004
tkritzer : 7/21/2004
terry : 7/16/2004
carol : 4/28/2004
tkritzer : 4/20/2004
mgross : 3/17/2004
cwells : 2/17/2004
terry : 2/9/2004
alopez : 11/7/2003
mgross : 10/30/2003
tkritzer : 10/7/2003
alopez : 8/7/2003
terry : 8/5/2003
mgross : 1/23/2003
mgross : 1/22/2003
alopez : 7/25/2002
mgross : 6/11/2002
mgross : 4/29/2002
mgross : 4/29/2002
mgross : 4/15/2002
mgross : 8/23/2000
mgross : 1/19/2000
alopez : 4/1/1999
carol : 11/2/1993
supermim : 3/16/1992
carol : 1/21/1992
supermim : 3/20/1990
ddp : 10/26/1989
root : 3/13/1989