Entry - *139311 - GUANINE NUCLEOTIDE-BINDING PROTEIN, ALPHA-ACTIVATING ACTIVITY POLYPEPTIDE O; GNAO1 - OMIM
 
* 139311

GUANINE NUCLEOTIDE-BINDING PROTEIN, ALPHA-ACTIVATING ACTIVITY POLYPEPTIDE O; GNAO1


Alternative titles; symbols

Go, ALPHA SUBUNIT
GUANINE NUCLEOTIDE-BINDING PROTEIN, ALPHA-'OTHER'; GNAO
G-ALPHA-o


HGNC Approved Gene Symbol: GNAO1

Cytogenetic location: 16q13     Genomic coordinates (GRCh38): 16:56,191,489-56,357,444 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
16q13 Developmental and epileptic encephalopathy 17 615473 AD 3
Neurodevelopmental disorder with involuntary movements 617493 AD 3

TEXT

Description

The GNAO1 gene encodes an alpha subunit of the heterotrimeric guanine nucleotide-binding proteins (G proteins), a large family of signal-transducing molecules. G proteins are composed of alpha, beta, and gamma subunits. Members of the G protein family have been characterized most extensively on the basis of the alpha subunit, which binds guanine nucleotide, is capable of hydrolyzing GTP, and interacts with specific receptor and effector molecules. In addition to the inhibitory G proteins, Gi (139310), and the stimulatory G proteins, Gs (139320), a Go protein has been described; 'o' means 'other.' The Go heterotrimer is abundant in brain and has been found also in the cardiac atria (summary by Strathmann et al., 1990).


Cloning and Expression

Strathmann et al. (1990) isolated cDNA clones encoding 2 forms of the Go-alpha subunit from a mouse brain library. These appear to be the products of alternative splicing. Tsukamoto et al. (1991) likewise concluded that 2 different Go-alpha mRNAs may be generated by alternative splicing of a single gene. Go-alpha has been implicated in ion channel regulation. Some tissues contain multiple Go-alpha mRNAs of various sizes that differ in the 3-prime untranslated regions (UTRs). Murtagh et al. (1991) concluded that the Go-alpha mRNAs with different 3-prime UTRs arise by alternative splicing of transcripts from a single gene. The UTRs were shown to exhibit a high degree of interspecies conservation and may play a role in regulation of Go-alpha expression during differentiation or in specific tissues.


Mapping

Murtagh et al. (1991) assigned the GNAO gene to chromosome 16 by Southern blot analysis of human-mouse somatic cell hybrids.

Stumpf (2020) mapped the GNAO1 gene to chromosome 16q13 based on an alignment of the GNAO1 sequence (GenBank BC030027) with the genomic sequence (GRCh38).

By study of an interspecific backcross with restriction fragment length variants (RFLVs), Wilkie et al. (1992) showed that the Gnao gene is located on mouse chromosome 8, where it is very tightly linked to the metallothionein gene (MT1; 156350).


Gene Function

In a bacterial expression system, Lan et al. (1998) found that point mutations in the Gnai1 and Gnao1 genes, G183S and G184S, respectively, resulted in resistance to regulators of G protein signaling proteins (RGS). The mutant G-alpha proteins showed significantly decreased affinity for RGS4 (602516) and RGS7 (602517).

Kroll et al. (1992) demonstrated that expression of Q205L Go-alpha, which lacks guanosine triphosphatase activity in NIH 3T3 cells, results in transformation in a phospholipase C (see 600220)-independent manner. Ram et al. (2000) studied the roles of the MAP kinases (see MAPK1, 176948) and STAT3 (102582) in transformation of NIH 3T3 cells by Q205L Go-alpha. Expression of Q205L Go-alpha in NIH 3T3 cells activated STAT3 but not MAPK1 or -2. Coexpression of dominant-negative STAT3 inhibited Q205L Go-alpha-induced transformation of NIH 3T3 cells and activation of endogenous STAT3. Furthermore, Q205L Go-alpha expression increased activity of the c-Src (190090), and the Q205L Go-alpha-induced activation of STAT3 was blocked by expression of CSK (124095), which inactivates c-Src. Ram et al. (2000) concluded that STAT3 can function as a downstream effector for Q205L Go-alpha and mediate its biologic effects.

Kan et al. (2010) reported the identification of 2,576 somatic mutations across approximately 1,800 megabases of DNA representing 1,507 coding genes from 441 tumors comprising breast, lung, ovarian, and prostate cancer types and subtypes. Integrated analysis of somatic mutations and copy number alterations identified 35 significantly altered genes including GNAS (see 139320), indicating an expanded role for G-alpha subunits in multiple cancer types. Experimental analyses demonstrated the functional roles of mutant GNAO1 and mutant MAP2K4 (601335) in oncogenesis.


Molecular Genetics

Developmental and Epileptic Encephalopathy 17

In 4 unrelated girls with developmental and epileptic encephalopathy-17 (DEE17; 615473), Nakamura et al. (2013) identified 4 different de novo heterozygous mutations in the GNAO1 gene (139311.0001-139311.0004). The mutations in the first 2 patients were found by whole-exome sequencing, and the mutations in the second 2 patients were found by direct sequencing of the GNAO1 gene in 367 individuals with epileptic encephalopathy. Three patients had onset of intractable tonic seizures in the first weeks of life associated with suppression-burst pattern on EEG, consistent with a clinical diagnosis of Ohtahara syndrome. The fourth patient presented with opisthotonic posturing and developmental delay at age 7 months. All had severely delayed psychomotor development, with lack of sitting, no speech, and head control only in 1 patient. One child died at age 11 months. One patient showed dystonia and another had severe chorea and athetosis. Brain MRI was abnormal in 3 patients, showing cerebral atrophy, delayed myelination, and thin corpus callosum. In vitro functional expression studies showed that 3 of the mutations impaired normal protein localization in the plasma membrane, and electrophysiologic analysis showed that 3 of the mutations caused decreased GNAO1-mediated inhibition of calcium currents by norepinephrine compared to wildtype. The findings suggested that aberrant GNAO1 signaling can cause multiple neurodevelopmental phenotypes, including epileptic encephalopathy and involuntary movements.

Neurodevelopmental Disorder With Involuntary Movements

In 2 brothers with neurodevelopmental disorder with involuntary movements (NEDIM; 617493), Kulkarni et al. (2016) identified a de novo heterozygous missense mutation in the GNAO1 gene (R209H; 139311.0005). The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in either parent, suggesting germline mosaicism in 1 of them. Functional studies of the variant were not performed, but it was predicted to disrupt GNAO1 signaling. Using exome sequencing, Menke et al. (2016) identified a de novo heterozygous R209H mutation in a 3-year-old boy with NEDIM. Functional studies of the variant were not performed.

In 2 unrelated patients with NEDIM, Saitsu et al. (2016) identified 2 different de novo heterozygous missense mutations in the GNAO1 gene (R209C, 139311.0006 and E246K, 139311.0007). The mutations were found by whole-exome sequencing and confirmed by Sanger sequencing. Functional studies of the variant and studies of patient cells were not performed, but molecular modeling predicted that they would result in adverse effects.

In 6 patients, including 2 sibs, with NEDIM, Ananth et al. (2016) identified de novo heterozygous missense mutations in the GNAO1 gene: E246K was found in 4 patients, R209H was found in 1 patient, and R209G (139311.0008) was found in 1 patient. The mutations were found by whole-exome sequencing. Functional studies of the variants and studies of patient cells were not performed. None of the patients had seizures, suggesting that these mutations may be specific to the movement disorder.

In 6 patients with NEDIM, Danti et al. (2017) identified de novo heterozygous mutations in the GNAO1 gene (see, e.g., R209C, 139311.0006 and E246G, 139311.0009).


Genotype/Phenotype Correlations

In 2 unrelated boys with neurodevelopmental disorder with involuntary movements (NEDIM; 617493), Menke et al. (2016) identified de novo heterozygous missense mutations affecting codon 209 in the GNAO1 gene (R209H, 139311.0005 and R209L). In a review of 26 published patients with GNAO1 mutations, Menke et al. (2016) found that those with mutations affecting codon 209 (e.g., R209C, R209H, R209G) or 246 (E246K; 139311.0007) had developmental delay with a hyperkinetic movement disorder but without seizures. These mutations were recurrent de novo mutations, probably related to both being part of CpG dinucleotides, which are known to be vulnerable to spontaneous deamination. In contrast, patients with mutations affecting other residues had the more severe phenotype of infantile-onset epileptic encephalopathy (DEE17). Menke et al. (2016) noted that affected sib pairs with the same de novo mutation had been reported, and they estimated a recurrence risk of 5 to 15% after 1 affected child with GNAO1 mutations.

Feng et al. (2017) performed functional and biochemical studies of 15 de novo GNAO1 mutations. Western blot analysis of HEK293 cells showed that most of the mutations resulted in decreased protein levels. Three variants affecting Arg209 showed normal protein expression, as did G184S. Functional studies assessing GNAO1-dependent cAMP inhibition when coexpressed with an adrenergic receptor showed that 9 of the mutations resulted in a loss of function (LOF), usually associated with significantly decreased protein levels, whereas 6 had normal or even gain-of-function (GOF) behavior compared to wildtype. The LOF variants were associated with DEE17, whereas the normal or GOF variants were associated with movement disorders with or without seizures. Molecular modeling also showed some correlation with the location of the mutations: GOF mutations were near G184S and close to the ribose and phosphate moieties of the bound GDP, whereas LOF mutations were more broadly scattered throughout the GTPase domain and may destabilize protein folding or stability consistent with their markedly reduced expression levels. Feng et al. (2017) discussed the possible therapeutic implications of their findings.


Animal Model

Jiang et al. (1998) disrupted the Gnao1 gene in mice by homologous recombination; median survival was only 7 weeks. At the cellular level, inhibition of cardiac adenylyl cyclase by carbachol was unaffected, but opioid receptor-mediated inhibition of calcium channel currents was decreased by 30%. In 25% of the homozygous mutant cells examined, the calcium channel was activated at voltages that were 13.3 +/- 1.7 mV lower than in their counterparts. Loss of alpha-o was not accompanied by appearance of significant amounts of active free beta-gamma dimers. Homozygous mutant mice were hyperalgesic and displayed a severe motor control impairment. Despite this problem, homozygous mutant mice were hyperactive and exhibited a turning behavior that had them running in circles for hours on end both in cages and in open-field tests. Except for one, all mutant mice turned counterclockwise. These results indicate that Go plays a major role in motor control, motor behavior, and pain perception and predict involvement of Go in calcium channel regulation.

To analyze the function of Go-alpha in the heart, Valenzuela et al. (1997) generated knockout mice lacking both forms of Go-alpha by homologous recombination and studied the muscarinic regulation of calcium channels in cardiac muscles in Go-alpha -/- mice and controls. There was no difference in the effect of isoproterenol on the L-type voltage-dependent calcium channel (114205) in ventricular myocytes of both groups, but the inhibitory effect of carbamylcholine was almost completely abolished in the Go-alpha -/- group. This demonstrated that, in the heart, Go-alpha is specifically required for transmission of signals from the muscarinic receptor to the L-type voltage-dependent calcium channel.

Go-alpha has been implicated as the primary signaling element coupling alpha-2-adrenergic receptors to N-type calcium channels in sympathetic neurons. Jeong and Ikeda (2000) found that in rat neurons expressing a Go-alpha subunit resistant to pertussis toxin and resistant to regulators of G protein signaling proteins, norepinephrine-induced calcium current inhibition was shifted to lower concentrations. In addition to an increase in agonist potency, the expression of the resistant Go-alpha subunit retarded the current recovery after agonist removal. The data suggested that endogenous RGS proteins contribute to calcium channel modulation by regulating agonist potency and kinetics of G protein-mediated signaling in neuronal cells.

Kehrl et al. (2014) found that mutant mice heterozygous for a G184S mutation in the Gnao1 gene died in the perinatal period or early in life due to sudden death associated with severe seizures and/or increased frequency of interictal epileptiform discharges. Homozygous mutant mice were essentially nonviable. Heterozygous mice showed enhanced sensitivity to seizure kindling with a GABA antagonist compared to controls. Heterozygous knockout mice, representing a loss of function, did not show such a phenotype, suggesting that the G184S mutation results in a gain of function. Kehrl et al. (2014) noted that several studies have shown that the G184S allele results in a gain of function effect.


ALLELIC VARIANTS ( 9 Selected Examples):

.0001 DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 17

GNAO1, ILE279ASN
  
RCV000056405

In a 13-year-old girl with developmental and epileptic encephalopathy-17 (DEE17; 615473), Nakamura et al. (2013) identified a de novo heterozygous c.836T-A transversion in the GNAO1 gene, resulting in an ile279-to-asn (I279N) substitution. The mutation specifically affected GNAO1 transcript variant 1. The mutation was found by whole-exome sequencing and was not found in the NHLBI Exome Sequencing Project database or in 408 in-house control exomes. In vitro functional expression studies in N2A cells showed that the mutant protein had some abnormal cytoplasmic localization. The patient had onset of seizures on day 4 of life. EEG showed a burst-suppression pattern consistent with a clinical diagnosis of Ohtahara syndrome.


.0002 DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 17

GNAO1, ASP174GLY
  
RCV000056406...

In a 4-year-old girl with developmental and epileptic encephalopathy-17 (DEE17; 615473), Nakamura et al. (2013) identified a de novo heterozygous c.521A-G transition in the GNAO1 gene, resulting in an asp174-to-gly (D174G) substitution. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was determined to be somatic mosaic by deep sequencing of PCR products of DNA from blood and saliva samples from the patient and her parents. The mutation was not found in the NHLBI Exome Sequencing Project database or in 408 in-house control exomes. In vitro functional expression studies in N2A cells showed that the mutant protein had some abnormal cytoplasmic localization. Electrophysiologic studies in N-type calcium channels indicated that the mutant protein had impaired current inhibition after norepinephrine application compared to wildtype, suggesting that the mutation could hamper GNAO1-mediated signaling. The patient had onset of seizures at 29 days of age. EEG showed a burst-suppression pattern consistent with a clinical diagnosis of Ohtahara syndrome.


.0003 DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 17

GNAO1, 21-BP DEL, NT572
  
RCV000056407

In a female infant with developmental and epileptic encephalopathy-17 (DEE17; 615473), Nakamura et al. (2013) identified a de novo identified a de novo heterozygous 21-bp deletion (c.572_592del), resulting in an in-frame deletion of 7 residues (Thr191_Phe197). The mutation was not found in the NHLBI Exome Sequencing Project database or in 408 in-house control exomes. In vitro functional expression studies in N2A cells showed that the mutant protein accumulated in the cytoplasmic compartment instead of being normally located to the cell periphery. Electrophysiologic studies in N-type calcium channels showed that the mutant protein had increased calcium-current density compared to wildtype before norepinephrine application, and showed only a mild reduction in calcium current compared to wildtype after application of norepinephrine. The findings suggested that the mutation could hamper GNAO1-mediated signaling. The patient had onset of intractable seizures at 2 weeks of age. EEG showed a burst-suppression pattern consistent with a clinical diagnosis of Ohtahara syndrome. She died at 11 months of age.


.0004 DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 17

GNAO1, GLY203ARG
   RCV000056408...

In an 8-year-old girl with developmental and epileptic encephalopathy-17 (DEE17; 615473), Nakamura et al. (2013) identified a de novo heterozygous c.607G-A transition in the GNAO1 gene, resulting in a gly203-to-arg (G203R) substitution in the highly conserved switch II region that is responsible for activation of downstream effectors. The mutation was not found in the NHLBI Exome Sequencing Project database or in 408 in-house control exomes. In vitro functional expression studies in N2A cells showed that the mutant protein localized normally to the cell periphery. However, electrophysiologic studies in N-type calcium channels indicated that the similar G203T mutant protein had impaired current inhibition after norepinephrine application compared to wildtype, suggesting that the mutation could hamper GNAO1-mediated signaling. The patient showed opisthotonic posturing at 7 months of age. She later developed severe chorea.

In a 14-month-old girl with DEE17, Saitsu et al. (2016) identified a de novo heterozygous G203R mutation in the GNAO1 gene. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the Exome Variant Server database or in 575 in-house control exomes. Functional studies of the variant and studies of patient cells were not performed. The patient developed seizures on the seventh day of life. She later developed severe chorea.

Feng et al. (2017) found that the G203R variant resulted in a gain-of-function effect in a cAMP inhibition assay. The authors noted that the previously reported patients with this variant had a slightly different phenotype from classic DEE17, showing a prominent motor component.


.0005 NEURODEVELOPMENTAL DISORDER WITH INVOLUNTARY MOVEMENTS

GNAO1, ARG209HIS
  
RCV000190691...

In 2 brothers with neurodevelopmental disorder with involuntary movements (NEDIM; 617493), Kulkarni et al. (2016) identified a de novo heterozygous c.626G-A transition in exon 6 of the GNAO1 gene, resulting in an arg209-to-his (R209H) substitution at a conserved residue in the highly conserved switch II region, which activates downstream effectors upon GTP binding. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in either parent, suggesting germline mosaicism in one of them. The mutation was filtered against the dbSNP and 1000 Genomes Project databases and was not found in the Exome Sequencing Project database. Functional studies of the variant were not performed, but it was predicted to disrupt GNAO1 signaling.

Using exome sequencing, Menke et al. (2016) identified a de novo heterozygous R209H mutation in a 3-year-old boy with NEDIM. Functional studies of the variant were not performed.

In a 16-year-old boy with NEDIM, Ananth et al. (2016) identified a de novo heterozygous R209H mutation in the GNAO1 gene. The mutation was found by whole-exome sequencing. Functional studies of the variant and studies of patient cells were not performed.


.0006 NEURODEVELOPMENTAL DISORDER WITH INVOLUNTARY MOVEMENTS

GNAO1, ARG209CYS
  
RCV000256155...

In an 18-year-old female with neurodevelopmental disorder with involuntary movements (NEDIM; 617493), Saitsu et al. (2016) identified a de novo heterozygous c.625C-T transition (c.625C-T, NM_020988.2) in exon 6 of the GNAO1 gene, resulting in an arg209-to-cys (R209C) substitution at a highly conserved residue. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the Exome Variant Server database or in 575 in-house control exomes. Functional studies of the variant and studies of patient cells were not performed, but molecular modeling predicted that the mutation would destabilize the G-alpha-containing complexes mainly in GTP-bound active state.

Danti et al. (2017) identified a de novo heterozygous R209C mutation in 2 unrelated patients with NEDIM. The mutation was not found in the 1000 Genomes Project, Exome Variant Server, or ExAC databases. Danti et al. (2017) noted that the R209C mutation occurs in the switch II domain, which is important for regulation of downstream signaling. This residue (R209) is a mutational hotspot.


.0007 NEURODEVELOPMENTAL DISORDER WITH INVOLUNTARY MOVEMENTS

GNAO1, GLU246LYS
  
RCV000190803...

In a 13-year-old girl with neurodevelopmental disorder with involuntary movements (NEDIM; 617493), Saitsu et al. (2016) identified a de novo heterozygous c.736G-A transition (c.736G-A, NM_020988.2) in exon 7 of the GNAO1 gene, resulting in a glu246-to-lys (E246K) substitution at a highly conserved residue. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the Exome Variant Server database or in 575 in-house control exomes. Functional studies of the variant and studies of patient cells were not performed, but molecular modeling predicted that the mutation would destabilize the G-alpha-containing complexes mainly in GTP-bound active state.

In 4 patients, including 2 sibs, with NEDIM, Ananth et al. (2016) identified a de novo heterozygous E246K mutation. The mutations were found by whole-exome sequencing. Functional studies of the variants and studies of patient cells were not performed.


.0008 NEURODEVELOPMENTAL DISORDER WITH INVOLUNTARY MOVEMENTS

GNAO1, ARG209GLY
  
RCV000490630

In a 4-year-old girl with neurodevelopmental disorder with involuntary movements (NEDIM; 617493), Ananth et al. (2016) identified a de novo heterozygous c.625C-G transversion in the GNAO1 gene, resulting in an arg209-to-gly (R209G) substitution at a highly conserved residue. The mutation was found by whole-exome sequencing. Functional studies of the variant and studies of patient cells were not performed.


.0009 NEURODEVELOPMENTAL DISORDER WITH INVOLUNTARY MOVEMENTS

GNAO1, GLU246GLY
  
RCV000490634

In a patient with neurodevelopmental disorder with involuntary movements (NEDIM; 617493), Danti et al. (2017) identified a de novo heterozygous c.737A-G transition in the GNAO1 gene, resulting in a glu246-to-gly (E246G) substitution. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the 1000 Genomes Project, Exome Variant Server, or ExAC databases. Functional studies of the variant and studies of patient cells were not performed.


REFERENCES

  1. Ananth, A. L., Robichaux-Viehoever, A., Kim, Y.-M., Hanson-Kahn, A., Cox, R., Enns, G. M., Strober, J., Willing, M., Schlaggar, B. L., Wu, Y. W., Bernstein, J. A. Clinical course of six children with GNAO1 mutations causing a severe and distinctive movement disorder. Pediat. Neurol. 59: 81-84, 2016. [PubMed: 27068059, related citations] [Full Text]

  2. Danti, F. R., Galosi, S., Romani, M., Montomoli, M., Carss, K. J., Raymond, F. L., Parrini, E., Bianchini, C., McShane, T., Dale, R. C., Mohammad, S. S., Shah, U., and 9 others. GNAO1 encephalopathy: broadening the phenotype and evaluating treatment and outcome. Neurol. Genet. 3: e143, 2017. Note: Electronic Article. [PubMed: 28357411, images, related citations] [Full Text]

  3. Feng, H., Sjogren, B., Karaj, B., Shaw, V., Gezer, A., Neugib, R. R. Movement disorder in GNAO1 encephalopathy associated with gain-of-function mutations. Neurology 89: 762-770, 2017. [PubMed: 28747448, related citations] [Full Text]

  4. Jeong, S.-W., Ikeda, S. R. Endogenous regulator of G-protein signaling proteins modify N-type calcium channel modulation in rat sympathetic neurons. J. Neurosci. 20: 4489-4496, 2000. [PubMed: 10844018, related citations] [Full Text]

  5. Jiang, M., Gold, M. S., Boulay, G., Spicher, K., Peyton, M., Brabet, P., Srinivasan, Y., Rudolph, U., Ellison, G., Birnbaumer, L. Multiple neurological abnormalities in mice deficient in the G protein G(o). Proc. Nat. Acad. Sci. 95: 3269-3274, 1998. [PubMed: 9501252, images, related citations] [Full Text]

  6. Kan, Z., Jaiswal, B. S., Stinson, J., Janakiraman, V., Bhatt, D., Stern, H. M., Yue, P., Haverty, P. M., Bourgon, R., Zheng, J., Moorhead, M., Chaudhuri, S., and 20 others. Diverse somatic mutation patterns and pathway alterations in human cancers. Nature 466: 869-873, 2010. [PubMed: 20668451, related citations] [Full Text]

  7. Kehrl, J. M., Sahaya, K., Dalton, H. M., Charbeneau, R. A., Kohut, K. T., Gilbert, K., Pelz, M. C., Parent, J., Neubig, R. R. Gain-of-function mutation in Gnao1: a murine model of epileptiform encephalopathy (EIEE17)? Mammalian Genome 25: 202-210, 2014. [PubMed: 24700286, images, related citations] [Full Text]

  8. Kroll, S. D., Chen, J., De Vivo, M., Carty, D. J., Buku, A., Premont, R. T., Iyengar, R. The Q205L G(o)-alpha subunit expressed in NIH-3T3 cells induces transformation. J. Biol. Chem. 267: 23183-23188, 1992. [PubMed: 1429665, related citations]

  9. Kulkarni, N., Tang, S., Bhardwaj, R., Bernes, S., Grebe, T. A. Progressive movement disorder in brothers carrying a GNAO1 mutation responsive to deep brain stimulation. J. Child. Neurol. 31: 211-214, 2016. [PubMed: 26060304, related citations] [Full Text]

  10. Lan, K.-L., Sarvazyan, N. A., Taussig, R., Mackenzie, R. G., DiBello, P. R., Dohlman, H. G., Neubig, R. R. A point mutation in G-alpha-o and G-alpha-i1 blocks interaction with regulator of G protein signaling proteins. J. Biol. Chem. 273: 12794-12797, 1998. [PubMed: 9582306, related citations] [Full Text]

  11. Menke, L. A., Engelen, M., Alders, M., Odekerken, V. J. J., Baas, F., Cobben, J. M. Recurrent GNAO1 mutations associated with developmental delay and a movement disorder. J. Child Neurol. 31: 1598-1601, 2016. [PubMed: 27625011, related citations] [Full Text]

  12. Murtagh, J. J., Jr., Eddy, R., Shows, T. B., Moss, J., Vaughan, M. Different forms of Go alpha mRNA arise by alternative splicing of transcripts from a single gene on human chromosome 16. Molec. Cell. Biol. 11: 1146-1155, 1991. [PubMed: 1899283, related citations] [Full Text]

  13. Nakamura, K., Kodera, H., Akita, T., Shiina, M., Kato, M., Hoshino, H., Terashima, H., Osaka, H., Nakamura, S., Tohyama, J., Kumada, T., Furukawa, T., and 14 others. De novo mutations in GNAO1, encoding a G-alpha-o subunit of heterotrimeric G proteins, cause epileptic encephalopathy. Am. J. Hum. Genet. 93: 496-505, 2013. [PubMed: 23993195, images, related citations] [Full Text]

  14. Ram, P. T., Horvath, C. M., Iyengar, R. Stat3-mediated transformation of NIH-3T3 cells by the constitutively active Q205L G-alpha(o) protein. Science 287: 142-144, 2000. [PubMed: 10615050, related citations] [Full Text]

  15. Saitsu, H., Fukai, R., Ben-Zeev, B., Sakai, Y., Mimaki, M., Okamoto, N., Suzuki, Y., Monden, Y., Saito, H., Tziperman, B., Torio, M., Akamine, S., and 10 others. Phenotypic spectrum of GNAO1 variants: epileptic encephalopathy to involuntary movements with severe developmental delay. Europ. J. Hum. Genet. 24: 129-134, 2016. [PubMed: 25966631, images, related citations] [Full Text]

  16. Strathmann, M., Wilkie, T. M., Simon, M. I. Alternative splicing produces transcripts encoding two forms of the alpha subunit of GTP-binding protein G(o). Proc. Nat. Acad. Sci. 87: 6477-6481, 1990. [PubMed: 1697681, related citations] [Full Text]

  17. Stumpf, A. M. Personal Communication. Baltimore, Md. 10/21/2020.

  18. Tsukamoto, T., Toyama, R., Itoh, H., Kozasa, T., Matsuoka, M., Kaziro, Y. Structure of the human gene and two rat cDNAs encoding the alpha chain of GTP-binding regulatory protein G(o): two different mRNAs are generated by alternative splicing. Proc. Nat. Acad. Sci. 88: 2974-2978, 1991. [PubMed: 1901650, related citations] [Full Text]

  19. Valenzuela, D., Han, X., Mende, U., Fankhauser, C., Mashimo, H., Huang, P., Pfeffer, J., Neer, E. J., Fishman, M. C. G-alpha-o is necessary for muscarinic regulation of Ca(2+) channels in mouse heart. Proc. Nat. Acad. Sci. 94: 1727-2732, 1997. [PubMed: 9050846, images, related citations] [Full Text]

  20. Wilkie, T. M., Gilbert, D. J., Olsen, A. S., Chen, X.-N., Amatruda, T. T., Korenberg, J. R., Trask, B. J., de Jong, P., Reed, R. R., Simon, M. I., Jenkins, N. A., Copeland, N. G. Evolution of the mammalian G protein alpha subunit multigene family. Nature Genet. 1: 85-91, 1992. [PubMed: 1302014, related citations] [Full Text]


Anne M. Stumpf - updated : 10/21/2020
Cassandra L. Kniffin - updated : 02/27/2018
Cassandra L. Kniffin - updated : 05/31/2017
Cassandra L. Kniffin - updated : 9/16/2015
Cassandra L. Kniffin - updated : 10/15/2013
Ada Hamosh - updated : 9/21/2010
Cassandra L. Kniffin - updated : 6/5/2006
Ada Hamosh - updated : 12/30/1999
Wilson H. Y. Lo - updated : 7/26/1999
Ada Hamosh - updated : 3/18/1999
Creation Date:
Victor A. McKusick : 10/16/1990
alopez : 10/21/2020
joanna : 10/09/2020
carol : 03/08/2018
ckniffin : 02/27/2018
carol : 06/02/2017
ckniffin : 05/31/2017
carol : 09/18/2015
carol : 9/17/2015
ckniffin : 9/16/2015
carol : 10/16/2013
ckniffin : 10/15/2013
alopez : 9/23/2010
terry : 9/21/2010
carol : 6/29/2006
ckniffin : 6/5/2006
alopez : 6/13/2001
alopez : 12/30/1999
carol : 7/26/1999
alopez : 3/18/1999
alopez : 3/18/1999
carol : 7/2/1998
mark : 9/3/1997
carol : 7/6/1992
carol : 5/26/1992
carol : 5/19/1992
supermim : 3/16/1992
carol : 5/13/1991
carol : 10/16/1990

* 139311

GUANINE NUCLEOTIDE-BINDING PROTEIN, ALPHA-ACTIVATING ACTIVITY POLYPEPTIDE O; GNAO1


Alternative titles; symbols

Go, ALPHA SUBUNIT
GUANINE NUCLEOTIDE-BINDING PROTEIN, ALPHA-'OTHER'; GNAO
G-ALPHA-o


HGNC Approved Gene Symbol: GNAO1

Cytogenetic location: 16q13     Genomic coordinates (GRCh38): 16:56,191,489-56,357,444 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
16q13 Developmental and epileptic encephalopathy 17 615473 Autosomal dominant 3
Neurodevelopmental disorder with involuntary movements 617493 Autosomal dominant 3

TEXT

Description

The GNAO1 gene encodes an alpha subunit of the heterotrimeric guanine nucleotide-binding proteins (G proteins), a large family of signal-transducing molecules. G proteins are composed of alpha, beta, and gamma subunits. Members of the G protein family have been characterized most extensively on the basis of the alpha subunit, which binds guanine nucleotide, is capable of hydrolyzing GTP, and interacts with specific receptor and effector molecules. In addition to the inhibitory G proteins, Gi (139310), and the stimulatory G proteins, Gs (139320), a Go protein has been described; 'o' means 'other.' The Go heterotrimer is abundant in brain and has been found also in the cardiac atria (summary by Strathmann et al., 1990).


Cloning and Expression

Strathmann et al. (1990) isolated cDNA clones encoding 2 forms of the Go-alpha subunit from a mouse brain library. These appear to be the products of alternative splicing. Tsukamoto et al. (1991) likewise concluded that 2 different Go-alpha mRNAs may be generated by alternative splicing of a single gene. Go-alpha has been implicated in ion channel regulation. Some tissues contain multiple Go-alpha mRNAs of various sizes that differ in the 3-prime untranslated regions (UTRs). Murtagh et al. (1991) concluded that the Go-alpha mRNAs with different 3-prime UTRs arise by alternative splicing of transcripts from a single gene. The UTRs were shown to exhibit a high degree of interspecies conservation and may play a role in regulation of Go-alpha expression during differentiation or in specific tissues.


Mapping

Murtagh et al. (1991) assigned the GNAO gene to chromosome 16 by Southern blot analysis of human-mouse somatic cell hybrids.

Stumpf (2020) mapped the GNAO1 gene to chromosome 16q13 based on an alignment of the GNAO1 sequence (GenBank BC030027) with the genomic sequence (GRCh38).

By study of an interspecific backcross with restriction fragment length variants (RFLVs), Wilkie et al. (1992) showed that the Gnao gene is located on mouse chromosome 8, where it is very tightly linked to the metallothionein gene (MT1; 156350).


Gene Function

In a bacterial expression system, Lan et al. (1998) found that point mutations in the Gnai1 and Gnao1 genes, G183S and G184S, respectively, resulted in resistance to regulators of G protein signaling proteins (RGS). The mutant G-alpha proteins showed significantly decreased affinity for RGS4 (602516) and RGS7 (602517).

Kroll et al. (1992) demonstrated that expression of Q205L Go-alpha, which lacks guanosine triphosphatase activity in NIH 3T3 cells, results in transformation in a phospholipase C (see 600220)-independent manner. Ram et al. (2000) studied the roles of the MAP kinases (see MAPK1, 176948) and STAT3 (102582) in transformation of NIH 3T3 cells by Q205L Go-alpha. Expression of Q205L Go-alpha in NIH 3T3 cells activated STAT3 but not MAPK1 or -2. Coexpression of dominant-negative STAT3 inhibited Q205L Go-alpha-induced transformation of NIH 3T3 cells and activation of endogenous STAT3. Furthermore, Q205L Go-alpha expression increased activity of the c-Src (190090), and the Q205L Go-alpha-induced activation of STAT3 was blocked by expression of CSK (124095), which inactivates c-Src. Ram et al. (2000) concluded that STAT3 can function as a downstream effector for Q205L Go-alpha and mediate its biologic effects.

Kan et al. (2010) reported the identification of 2,576 somatic mutations across approximately 1,800 megabases of DNA representing 1,507 coding genes from 441 tumors comprising breast, lung, ovarian, and prostate cancer types and subtypes. Integrated analysis of somatic mutations and copy number alterations identified 35 significantly altered genes including GNAS (see 139320), indicating an expanded role for G-alpha subunits in multiple cancer types. Experimental analyses demonstrated the functional roles of mutant GNAO1 and mutant MAP2K4 (601335) in oncogenesis.


Molecular Genetics

Developmental and Epileptic Encephalopathy 17

In 4 unrelated girls with developmental and epileptic encephalopathy-17 (DEE17; 615473), Nakamura et al. (2013) identified 4 different de novo heterozygous mutations in the GNAO1 gene (139311.0001-139311.0004). The mutations in the first 2 patients were found by whole-exome sequencing, and the mutations in the second 2 patients were found by direct sequencing of the GNAO1 gene in 367 individuals with epileptic encephalopathy. Three patients had onset of intractable tonic seizures in the first weeks of life associated with suppression-burst pattern on EEG, consistent with a clinical diagnosis of Ohtahara syndrome. The fourth patient presented with opisthotonic posturing and developmental delay at age 7 months. All had severely delayed psychomotor development, with lack of sitting, no speech, and head control only in 1 patient. One child died at age 11 months. One patient showed dystonia and another had severe chorea and athetosis. Brain MRI was abnormal in 3 patients, showing cerebral atrophy, delayed myelination, and thin corpus callosum. In vitro functional expression studies showed that 3 of the mutations impaired normal protein localization in the plasma membrane, and electrophysiologic analysis showed that 3 of the mutations caused decreased GNAO1-mediated inhibition of calcium currents by norepinephrine compared to wildtype. The findings suggested that aberrant GNAO1 signaling can cause multiple neurodevelopmental phenotypes, including epileptic encephalopathy and involuntary movements.

Neurodevelopmental Disorder With Involuntary Movements

In 2 brothers with neurodevelopmental disorder with involuntary movements (NEDIM; 617493), Kulkarni et al. (2016) identified a de novo heterozygous missense mutation in the GNAO1 gene (R209H; 139311.0005). The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in either parent, suggesting germline mosaicism in 1 of them. Functional studies of the variant were not performed, but it was predicted to disrupt GNAO1 signaling. Using exome sequencing, Menke et al. (2016) identified a de novo heterozygous R209H mutation in a 3-year-old boy with NEDIM. Functional studies of the variant were not performed.

In 2 unrelated patients with NEDIM, Saitsu et al. (2016) identified 2 different de novo heterozygous missense mutations in the GNAO1 gene (R209C, 139311.0006 and E246K, 139311.0007). The mutations were found by whole-exome sequencing and confirmed by Sanger sequencing. Functional studies of the variant and studies of patient cells were not performed, but molecular modeling predicted that they would result in adverse effects.

In 6 patients, including 2 sibs, with NEDIM, Ananth et al. (2016) identified de novo heterozygous missense mutations in the GNAO1 gene: E246K was found in 4 patients, R209H was found in 1 patient, and R209G (139311.0008) was found in 1 patient. The mutations were found by whole-exome sequencing. Functional studies of the variants and studies of patient cells were not performed. None of the patients had seizures, suggesting that these mutations may be specific to the movement disorder.

In 6 patients with NEDIM, Danti et al. (2017) identified de novo heterozygous mutations in the GNAO1 gene (see, e.g., R209C, 139311.0006 and E246G, 139311.0009).


Genotype/Phenotype Correlations

In 2 unrelated boys with neurodevelopmental disorder with involuntary movements (NEDIM; 617493), Menke et al. (2016) identified de novo heterozygous missense mutations affecting codon 209 in the GNAO1 gene (R209H, 139311.0005 and R209L). In a review of 26 published patients with GNAO1 mutations, Menke et al. (2016) found that those with mutations affecting codon 209 (e.g., R209C, R209H, R209G) or 246 (E246K; 139311.0007) had developmental delay with a hyperkinetic movement disorder but without seizures. These mutations were recurrent de novo mutations, probably related to both being part of CpG dinucleotides, which are known to be vulnerable to spontaneous deamination. In contrast, patients with mutations affecting other residues had the more severe phenotype of infantile-onset epileptic encephalopathy (DEE17). Menke et al. (2016) noted that affected sib pairs with the same de novo mutation had been reported, and they estimated a recurrence risk of 5 to 15% after 1 affected child with GNAO1 mutations.

Feng et al. (2017) performed functional and biochemical studies of 15 de novo GNAO1 mutations. Western blot analysis of HEK293 cells showed that most of the mutations resulted in decreased protein levels. Three variants affecting Arg209 showed normal protein expression, as did G184S. Functional studies assessing GNAO1-dependent cAMP inhibition when coexpressed with an adrenergic receptor showed that 9 of the mutations resulted in a loss of function (LOF), usually associated with significantly decreased protein levels, whereas 6 had normal or even gain-of-function (GOF) behavior compared to wildtype. The LOF variants were associated with DEE17, whereas the normal or GOF variants were associated with movement disorders with or without seizures. Molecular modeling also showed some correlation with the location of the mutations: GOF mutations were near G184S and close to the ribose and phosphate moieties of the bound GDP, whereas LOF mutations were more broadly scattered throughout the GTPase domain and may destabilize protein folding or stability consistent with their markedly reduced expression levels. Feng et al. (2017) discussed the possible therapeutic implications of their findings.


Animal Model

Jiang et al. (1998) disrupted the Gnao1 gene in mice by homologous recombination; median survival was only 7 weeks. At the cellular level, inhibition of cardiac adenylyl cyclase by carbachol was unaffected, but opioid receptor-mediated inhibition of calcium channel currents was decreased by 30%. In 25% of the homozygous mutant cells examined, the calcium channel was activated at voltages that were 13.3 +/- 1.7 mV lower than in their counterparts. Loss of alpha-o was not accompanied by appearance of significant amounts of active free beta-gamma dimers. Homozygous mutant mice were hyperalgesic and displayed a severe motor control impairment. Despite this problem, homozygous mutant mice were hyperactive and exhibited a turning behavior that had them running in circles for hours on end both in cages and in open-field tests. Except for one, all mutant mice turned counterclockwise. These results indicate that Go plays a major role in motor control, motor behavior, and pain perception and predict involvement of Go in calcium channel regulation.

To analyze the function of Go-alpha in the heart, Valenzuela et al. (1997) generated knockout mice lacking both forms of Go-alpha by homologous recombination and studied the muscarinic regulation of calcium channels in cardiac muscles in Go-alpha -/- mice and controls. There was no difference in the effect of isoproterenol on the L-type voltage-dependent calcium channel (114205) in ventricular myocytes of both groups, but the inhibitory effect of carbamylcholine was almost completely abolished in the Go-alpha -/- group. This demonstrated that, in the heart, Go-alpha is specifically required for transmission of signals from the muscarinic receptor to the L-type voltage-dependent calcium channel.

Go-alpha has been implicated as the primary signaling element coupling alpha-2-adrenergic receptors to N-type calcium channels in sympathetic neurons. Jeong and Ikeda (2000) found that in rat neurons expressing a Go-alpha subunit resistant to pertussis toxin and resistant to regulators of G protein signaling proteins, norepinephrine-induced calcium current inhibition was shifted to lower concentrations. In addition to an increase in agonist potency, the expression of the resistant Go-alpha subunit retarded the current recovery after agonist removal. The data suggested that endogenous RGS proteins contribute to calcium channel modulation by regulating agonist potency and kinetics of G protein-mediated signaling in neuronal cells.

Kehrl et al. (2014) found that mutant mice heterozygous for a G184S mutation in the Gnao1 gene died in the perinatal period or early in life due to sudden death associated with severe seizures and/or increased frequency of interictal epileptiform discharges. Homozygous mutant mice were essentially nonviable. Heterozygous mice showed enhanced sensitivity to seizure kindling with a GABA antagonist compared to controls. Heterozygous knockout mice, representing a loss of function, did not show such a phenotype, suggesting that the G184S mutation results in a gain of function. Kehrl et al. (2014) noted that several studies have shown that the G184S allele results in a gain of function effect.


ALLELIC VARIANTS 9 Selected Examples):

.0001   DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 17

GNAO1, ILE279ASN
SNP: rs587777054, ClinVar: RCV000056405

In a 13-year-old girl with developmental and epileptic encephalopathy-17 (DEE17; 615473), Nakamura et al. (2013) identified a de novo heterozygous c.836T-A transversion in the GNAO1 gene, resulting in an ile279-to-asn (I279N) substitution. The mutation specifically affected GNAO1 transcript variant 1. The mutation was found by whole-exome sequencing and was not found in the NHLBI Exome Sequencing Project database or in 408 in-house control exomes. In vitro functional expression studies in N2A cells showed that the mutant protein had some abnormal cytoplasmic localization. The patient had onset of seizures on day 4 of life. EEG showed a burst-suppression pattern consistent with a clinical diagnosis of Ohtahara syndrome.


.0002   DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 17

GNAO1, ASP174GLY
SNP: rs587777055, ClinVar: RCV000056406, RCV002281559

In a 4-year-old girl with developmental and epileptic encephalopathy-17 (DEE17; 615473), Nakamura et al. (2013) identified a de novo heterozygous c.521A-G transition in the GNAO1 gene, resulting in an asp174-to-gly (D174G) substitution. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was determined to be somatic mosaic by deep sequencing of PCR products of DNA from blood and saliva samples from the patient and her parents. The mutation was not found in the NHLBI Exome Sequencing Project database or in 408 in-house control exomes. In vitro functional expression studies in N2A cells showed that the mutant protein had some abnormal cytoplasmic localization. Electrophysiologic studies in N-type calcium channels indicated that the mutant protein had impaired current inhibition after norepinephrine application compared to wildtype, suggesting that the mutation could hamper GNAO1-mediated signaling. The patient had onset of seizures at 29 days of age. EEG showed a burst-suppression pattern consistent with a clinical diagnosis of Ohtahara syndrome.


.0003   DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 17

GNAO1, 21-BP DEL, NT572
SNP: rs587777056, ClinVar: RCV000056407

In a female infant with developmental and epileptic encephalopathy-17 (DEE17; 615473), Nakamura et al. (2013) identified a de novo identified a de novo heterozygous 21-bp deletion (c.572_592del), resulting in an in-frame deletion of 7 residues (Thr191_Phe197). The mutation was not found in the NHLBI Exome Sequencing Project database or in 408 in-house control exomes. In vitro functional expression studies in N2A cells showed that the mutant protein accumulated in the cytoplasmic compartment instead of being normally located to the cell periphery. Electrophysiologic studies in N-type calcium channels showed that the mutant protein had increased calcium-current density compared to wildtype before norepinephrine application, and showed only a mild reduction in calcium current compared to wildtype after application of norepinephrine. The findings suggested that the mutation could hamper GNAO1-mediated signaling. The patient had onset of intractable seizures at 2 weeks of age. EEG showed a burst-suppression pattern consistent with a clinical diagnosis of Ohtahara syndrome. She died at 11 months of age.


.0004   DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 17

GNAO1, GLY203ARG
ClinVar: RCV000056408, RCV000255097, RCV000468248, RCV000762963, RCV001252685, RCV001256978, RCV001814039, RCV003421966

In an 8-year-old girl with developmental and epileptic encephalopathy-17 (DEE17; 615473), Nakamura et al. (2013) identified a de novo heterozygous c.607G-A transition in the GNAO1 gene, resulting in a gly203-to-arg (G203R) substitution in the highly conserved switch II region that is responsible for activation of downstream effectors. The mutation was not found in the NHLBI Exome Sequencing Project database or in 408 in-house control exomes. In vitro functional expression studies in N2A cells showed that the mutant protein localized normally to the cell periphery. However, electrophysiologic studies in N-type calcium channels indicated that the similar G203T mutant protein had impaired current inhibition after norepinephrine application compared to wildtype, suggesting that the mutation could hamper GNAO1-mediated signaling. The patient showed opisthotonic posturing at 7 months of age. She later developed severe chorea.

In a 14-month-old girl with DEE17, Saitsu et al. (2016) identified a de novo heterozygous G203R mutation in the GNAO1 gene. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the Exome Variant Server database or in 575 in-house control exomes. Functional studies of the variant and studies of patient cells were not performed. The patient developed seizures on the seventh day of life. She later developed severe chorea.

Feng et al. (2017) found that the G203R variant resulted in a gain-of-function effect in a cAMP inhibition assay. The authors noted that the previously reported patients with this variant had a slightly different phenotype from classic DEE17, showing a prominent motor component.


.0005   NEURODEVELOPMENTAL DISORDER WITH INVOLUNTARY MOVEMENTS

GNAO1, ARG209HIS
SNP: rs797044878, ClinVar: RCV000190691, RCV000255659, RCV000490633, RCV001065368

In 2 brothers with neurodevelopmental disorder with involuntary movements (NEDIM; 617493), Kulkarni et al. (2016) identified a de novo heterozygous c.626G-A transition in exon 6 of the GNAO1 gene, resulting in an arg209-to-his (R209H) substitution at a conserved residue in the highly conserved switch II region, which activates downstream effectors upon GTP binding. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in either parent, suggesting germline mosaicism in one of them. The mutation was filtered against the dbSNP and 1000 Genomes Project databases and was not found in the Exome Sequencing Project database. Functional studies of the variant were not performed, but it was predicted to disrupt GNAO1 signaling.

Using exome sequencing, Menke et al. (2016) identified a de novo heterozygous R209H mutation in a 3-year-old boy with NEDIM. Functional studies of the variant were not performed.

In a 16-year-old boy with NEDIM, Ananth et al. (2016) identified a de novo heterozygous R209H mutation in the GNAO1 gene. The mutation was found by whole-exome sequencing. Functional studies of the variant and studies of patient cells were not performed.


.0006   NEURODEVELOPMENTAL DISORDER WITH INVOLUNTARY MOVEMENTS

GNAO1, ARG209CYS
SNP: rs886039494, ClinVar: RCV000256155, RCV000475848, RCV000490628, RCV000622320, RCV001003612, RCV001775107, RCV003401217

In an 18-year-old female with neurodevelopmental disorder with involuntary movements (NEDIM; 617493), Saitsu et al. (2016) identified a de novo heterozygous c.625C-T transition (c.625C-T, NM_020988.2) in exon 6 of the GNAO1 gene, resulting in an arg209-to-cys (R209C) substitution at a highly conserved residue. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the Exome Variant Server database or in 575 in-house control exomes. Functional studies of the variant and studies of patient cells were not performed, but molecular modeling predicted that the mutation would destabilize the G-alpha-containing complexes mainly in GTP-bound active state.

Danti et al. (2017) identified a de novo heterozygous R209C mutation in 2 unrelated patients with NEDIM. The mutation was not found in the 1000 Genomes Project, Exome Variant Server, or ExAC databases. Danti et al. (2017) noted that the R209C mutation occurs in the switch II domain, which is important for regulation of downstream signaling. This residue (R209) is a mutational hotspot.


.0007   NEURODEVELOPMENTAL DISORDER WITH INVOLUNTARY MOVEMENTS

GNAO1, GLU246LYS
SNP: rs797044951, ClinVar: RCV000190803, RCV000254701, RCV000490631, RCV001580372, RCV001808530, RCV001814097, RCV001857676, RCV003996903

In a 13-year-old girl with neurodevelopmental disorder with involuntary movements (NEDIM; 617493), Saitsu et al. (2016) identified a de novo heterozygous c.736G-A transition (c.736G-A, NM_020988.2) in exon 7 of the GNAO1 gene, resulting in a glu246-to-lys (E246K) substitution at a highly conserved residue. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the Exome Variant Server database or in 575 in-house control exomes. Functional studies of the variant and studies of patient cells were not performed, but molecular modeling predicted that the mutation would destabilize the G-alpha-containing complexes mainly in GTP-bound active state.

In 4 patients, including 2 sibs, with NEDIM, Ananth et al. (2016) identified a de novo heterozygous E246K mutation. The mutations were found by whole-exome sequencing. Functional studies of the variants and studies of patient cells were not performed.


.0008   NEURODEVELOPMENTAL DISORDER WITH INVOLUNTARY MOVEMENTS

GNAO1, ARG209GLY
SNP: rs886039494, ClinVar: RCV000490630

In a 4-year-old girl with neurodevelopmental disorder with involuntary movements (NEDIM; 617493), Ananth et al. (2016) identified a de novo heterozygous c.625C-G transversion in the GNAO1 gene, resulting in an arg209-to-gly (R209G) substitution at a highly conserved residue. The mutation was found by whole-exome sequencing. Functional studies of the variant and studies of patient cells were not performed.


.0009   NEURODEVELOPMENTAL DISORDER WITH INVOLUNTARY MOVEMENTS

GNAO1, GLU246GLY
SNP: rs1114167431, ClinVar: RCV000490634

In a patient with neurodevelopmental disorder with involuntary movements (NEDIM; 617493), Danti et al. (2017) identified a de novo heterozygous c.737A-G transition in the GNAO1 gene, resulting in a glu246-to-gly (E246G) substitution. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the 1000 Genomes Project, Exome Variant Server, or ExAC databases. Functional studies of the variant and studies of patient cells were not performed.


REFERENCES

  1. Ananth, A. L., Robichaux-Viehoever, A., Kim, Y.-M., Hanson-Kahn, A., Cox, R., Enns, G. M., Strober, J., Willing, M., Schlaggar, B. L., Wu, Y. W., Bernstein, J. A. Clinical course of six children with GNAO1 mutations causing a severe and distinctive movement disorder. Pediat. Neurol. 59: 81-84, 2016. [PubMed: 27068059] [Full Text: https://doi.org/10.1016/j.pediatrneurol.2016.02.018]

  2. Danti, F. R., Galosi, S., Romani, M., Montomoli, M., Carss, K. J., Raymond, F. L., Parrini, E., Bianchini, C., McShane, T., Dale, R. C., Mohammad, S. S., Shah, U., and 9 others. GNAO1 encephalopathy: broadening the phenotype and evaluating treatment and outcome. Neurol. Genet. 3: e143, 2017. Note: Electronic Article. [PubMed: 28357411] [Full Text: https://doi.org/10.1212/NXG.0000000000000143]

  3. Feng, H., Sjogren, B., Karaj, B., Shaw, V., Gezer, A., Neugib, R. R. Movement disorder in GNAO1 encephalopathy associated with gain-of-function mutations. Neurology 89: 762-770, 2017. [PubMed: 28747448] [Full Text: https://doi.org/10.1212/WNL.0000000000004262]

  4. Jeong, S.-W., Ikeda, S. R. Endogenous regulator of G-protein signaling proteins modify N-type calcium channel modulation in rat sympathetic neurons. J. Neurosci. 20: 4489-4496, 2000. [PubMed: 10844018] [Full Text: https://doi.org/10.1523/JNEUROSCI.20-12-04489.2000]

  5. Jiang, M., Gold, M. S., Boulay, G., Spicher, K., Peyton, M., Brabet, P., Srinivasan, Y., Rudolph, U., Ellison, G., Birnbaumer, L. Multiple neurological abnormalities in mice deficient in the G protein G(o). Proc. Nat. Acad. Sci. 95: 3269-3274, 1998. [PubMed: 9501252] [Full Text: https://doi.org/10.1073/pnas.95.6.3269]

  6. Kan, Z., Jaiswal, B. S., Stinson, J., Janakiraman, V., Bhatt, D., Stern, H. M., Yue, P., Haverty, P. M., Bourgon, R., Zheng, J., Moorhead, M., Chaudhuri, S., and 20 others. Diverse somatic mutation patterns and pathway alterations in human cancers. Nature 466: 869-873, 2010. [PubMed: 20668451] [Full Text: https://doi.org/10.1038/nature09208]

  7. Kehrl, J. M., Sahaya, K., Dalton, H. M., Charbeneau, R. A., Kohut, K. T., Gilbert, K., Pelz, M. C., Parent, J., Neubig, R. R. Gain-of-function mutation in Gnao1: a murine model of epileptiform encephalopathy (EIEE17)? Mammalian Genome 25: 202-210, 2014. [PubMed: 24700286] [Full Text: https://doi.org/10.1007/s00335-014-9509-z]

  8. Kroll, S. D., Chen, J., De Vivo, M., Carty, D. J., Buku, A., Premont, R. T., Iyengar, R. The Q205L G(o)-alpha subunit expressed in NIH-3T3 cells induces transformation. J. Biol. Chem. 267: 23183-23188, 1992. [PubMed: 1429665]

  9. Kulkarni, N., Tang, S., Bhardwaj, R., Bernes, S., Grebe, T. A. Progressive movement disorder in brothers carrying a GNAO1 mutation responsive to deep brain stimulation. J. Child. Neurol. 31: 211-214, 2016. [PubMed: 26060304] [Full Text: https://doi.org/10.1177/0883073815587945]

  10. Lan, K.-L., Sarvazyan, N. A., Taussig, R., Mackenzie, R. G., DiBello, P. R., Dohlman, H. G., Neubig, R. R. A point mutation in G-alpha-o and G-alpha-i1 blocks interaction with regulator of G protein signaling proteins. J. Biol. Chem. 273: 12794-12797, 1998. [PubMed: 9582306] [Full Text: https://doi.org/10.1074/jbc.273.21.12794]

  11. Menke, L. A., Engelen, M., Alders, M., Odekerken, V. J. J., Baas, F., Cobben, J. M. Recurrent GNAO1 mutations associated with developmental delay and a movement disorder. J. Child Neurol. 31: 1598-1601, 2016. [PubMed: 27625011] [Full Text: https://doi.org/10.1177/0883073816666474]

  12. Murtagh, J. J., Jr., Eddy, R., Shows, T. B., Moss, J., Vaughan, M. Different forms of Go alpha mRNA arise by alternative splicing of transcripts from a single gene on human chromosome 16. Molec. Cell. Biol. 11: 1146-1155, 1991. [PubMed: 1899283] [Full Text: https://doi.org/10.1128/mcb.11.2.1146-1155.1991]

  13. Nakamura, K., Kodera, H., Akita, T., Shiina, M., Kato, M., Hoshino, H., Terashima, H., Osaka, H., Nakamura, S., Tohyama, J., Kumada, T., Furukawa, T., and 14 others. De novo mutations in GNAO1, encoding a G-alpha-o subunit of heterotrimeric G proteins, cause epileptic encephalopathy. Am. J. Hum. Genet. 93: 496-505, 2013. [PubMed: 23993195] [Full Text: https://doi.org/10.1016/j.ajhg.2013.07.014]

  14. Ram, P. T., Horvath, C. M., Iyengar, R. Stat3-mediated transformation of NIH-3T3 cells by the constitutively active Q205L G-alpha(o) protein. Science 287: 142-144, 2000. [PubMed: 10615050] [Full Text: https://doi.org/10.1126/science.287.5450.142]

  15. Saitsu, H., Fukai, R., Ben-Zeev, B., Sakai, Y., Mimaki, M., Okamoto, N., Suzuki, Y., Monden, Y., Saito, H., Tziperman, B., Torio, M., Akamine, S., and 10 others. Phenotypic spectrum of GNAO1 variants: epileptic encephalopathy to involuntary movements with severe developmental delay. Europ. J. Hum. Genet. 24: 129-134, 2016. [PubMed: 25966631] [Full Text: https://doi.org/10.1038/ejhg.2015.92]

  16. Strathmann, M., Wilkie, T. M., Simon, M. I. Alternative splicing produces transcripts encoding two forms of the alpha subunit of GTP-binding protein G(o). Proc. Nat. Acad. Sci. 87: 6477-6481, 1990. [PubMed: 1697681] [Full Text: https://doi.org/10.1073/pnas.87.17.6477]

  17. Stumpf, A. M. Personal Communication. Baltimore, Md. 10/21/2020.

  18. Tsukamoto, T., Toyama, R., Itoh, H., Kozasa, T., Matsuoka, M., Kaziro, Y. Structure of the human gene and two rat cDNAs encoding the alpha chain of GTP-binding regulatory protein G(o): two different mRNAs are generated by alternative splicing. Proc. Nat. Acad. Sci. 88: 2974-2978, 1991. [PubMed: 1901650] [Full Text: https://doi.org/10.1073/pnas.88.8.2974]

  19. Valenzuela, D., Han, X., Mende, U., Fankhauser, C., Mashimo, H., Huang, P., Pfeffer, J., Neer, E. J., Fishman, M. C. G-alpha-o is necessary for muscarinic regulation of Ca(2+) channels in mouse heart. Proc. Nat. Acad. Sci. 94: 1727-2732, 1997. [PubMed: 9050846] [Full Text: https://doi.org/10.1073/pnas.94.5.1727]

  20. Wilkie, T. M., Gilbert, D. J., Olsen, A. S., Chen, X.-N., Amatruda, T. T., Korenberg, J. R., Trask, B. J., de Jong, P., Reed, R. R., Simon, M. I., Jenkins, N. A., Copeland, N. G. Evolution of the mammalian G protein alpha subunit multigene family. Nature Genet. 1: 85-91, 1992. [PubMed: 1302014] [Full Text: https://doi.org/10.1038/ng0592-85]


Contributors:
Anne M. Stumpf - updated : 10/21/2020
Cassandra L. Kniffin - updated : 02/27/2018
Cassandra L. Kniffin - updated : 05/31/2017
Cassandra L. Kniffin - updated : 9/16/2015
Cassandra L. Kniffin - updated : 10/15/2013
Ada Hamosh - updated : 9/21/2010
Cassandra L. Kniffin - updated : 6/5/2006
Ada Hamosh - updated : 12/30/1999
Wilson H. Y. Lo - updated : 7/26/1999
Ada Hamosh - updated : 3/18/1999

Creation Date:
Victor A. McKusick : 10/16/1990

Edit History:
alopez : 10/21/2020
joanna : 10/09/2020
carol : 03/08/2018
ckniffin : 02/27/2018
carol : 06/02/2017
ckniffin : 05/31/2017
carol : 09/18/2015
carol : 9/17/2015
ckniffin : 9/16/2015
carol : 10/16/2013
ckniffin : 10/15/2013
alopez : 9/23/2010
terry : 9/21/2010
carol : 6/29/2006
ckniffin : 6/5/2006
alopez : 6/13/2001
alopez : 12/30/1999
carol : 7/26/1999
alopez : 3/18/1999
alopez : 3/18/1999
carol : 7/2/1998
mark : 9/3/1997
carol : 7/6/1992
carol : 5/26/1992
carol : 5/19/1992
supermim : 3/16/1992
carol : 5/13/1991
carol : 10/16/1990