Entry - *600898 - SRY-BOX 11; SOX11 - OMIM
 
* 600898

SRY-BOX 11; SOX11


Alternative titles; symbols

SRY-RELATED HMG-BOX GENE 11


HGNC Approved Gene Symbol: SOX11

Cytogenetic location: 2p25.2     Genomic coordinates (GRCh38): 2:5,692,384-5,701,385 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
2p25.2 Intellectual developmental disorder with microcephaly and with or without ocular malformations or hypogonadotropic hypogonadism 615866 AD 3

TEXT

Cloning and Expression

Using the partial clones of both human and mouse SOX11 genes, Jay et al. (1995) cloned and characterized the human SOX11 gene. The SOX11 sequence is strongly conserved with the chicken homolog and is related to SOX4. It contains several putative transcriptional activator or repressor domains. The authors observed that the SOX11 expression pattern is consistent with the hypothesis that this gene is important in the developing nervous system.


Mapping

Jay et al. (1995) mapped the SOX11 gene to chromosome 2p25 by fluorescence in situ hybridization.


Gene Function

Shim et al. (2012) identified a conserved nonexonic element (E4), located 7.3 kb downstream of the Fezf2 (607414) transcription start site, that is required for the specification of corticospinal neuron identity and connectivity. Shim et al. (2012) found that Sox4 (184430) and Sox11 functionally compete with the repressor Sox5 (604975) in the transactivation of E4. Shim et al. (2012) showed that SOX4 and SOX11 are crucial in regulating reelin (RELN; 600514) expression and the inside-out pattern of cortical layer formation, independent of E4 or Fezf2 and probably involving interactions with distinct regulatory elements. Cortex-specific double deletion of Sox4 and Sox11 led to the loss of Fezf2 expression, failed specification of corticospinal neurons and, independent of Fezf2, a reeler-like inversion of layers. Moreover, SOX4 and SOX11 have additional roles, since in mice lacking both genes, the cortex and olfactory bulb are smaller and cell death is increased. Thus, SOX4 and SOX11 have pleiotropic functions, which are probably mediated by distinct regulatory elements and downstream target genes that are involved in multiple developmental processes. Shim et al. (2012) showed evidence supporting the emergence of functional SOX-binding sites in E4 during tetrapod evolution, and their subsequent stabilization in mammals and possibly amniotes. Shim et al. (2012) concluded that SOX transcription factors converge onto a cis-acting element of Fezf2 and form critical components of a regulatory network controlling the identity and connectivity of corticospinal neurons.


Biochemical Features

Cryoelectron Microscopy

Dodonova et al. (2020) reported cryoelectron microscopy structures of the DNA-binding domains of SOX2 (184429) and its close homolog SOX11 bound to nucleosomes. The structures showed that SOX factors can bind and locally distort DNA at superhelical location 2. The factors also facilitated detachment of terminal nucleosomal DNA from the histone octamer, which increases DNA accessibility. SOX-factor binding to the nucleosome can also lead to a repositioning of the N-terminal tail of histone H4 (see 602822) that includes residue lys16. Dodonova et al. (2020) speculated that this repositioning is incompatible with higher-order nucleosome stacking, which involves contacts of the H4 tail with a neighboring nucleosome. Dodonova et al. (2020) concluded that pioneer transcription factors that maintain pluripotency can use binding energy to initiate chromatin opening, and thereby facilitate nucleosome remodeling and subsequent transcription.


Gene Family

SRY (480000) is the testis-determining gene located on the Y chromosome of mammals. It encodes a protein whose most striking feature is a motif of 78 amino acids conserved with respect to the DNA binding domain of the high mobility group (HMG) proteins. Jay et al. (1995) noted that more than 100 HMG box-containing proteins had been reported at that time and are classified in 2 distinct subgroups according to the sequence-specificity of the binding, the number of DNA binding domains, and phylogenetic considerations (Laudet et al., 1993). An important subgroup of HMG box-containing proteins includes SRY and SRY box-related (SOX) sequences. They contain only 1 DNA-binding domain, and they bind to DNA in a sequence-specific manner. They are all potential transcription factors implicated in the developmental control of gene expression. Degenerate PCR-based methods enabled the cloning and sequencing of a great number of new SRY-related box sequences in both vertebrates and invertebrates.


Molecular Genetics

Tsurusaki et al. (2014) identified 2 de novo missense mutations in the SOX11 gene (Y116C, 600898.0001 and S60P, 600898.0002) in 2 unrelated female patients with intellectual developmental disorder with microcephaly and with or without ocular malformations or hypogonadotropic hypogonadism (IDDMOH; 615866), also referred to as Coffin-Siris syndrome-9 (CSS9). Both mutations occurred in the HMG domain in 2 evolutionarily conserved amino acids. Tsurusaki et al. (2014) showed that both mutations caused decreased transcriptional activation compared to wildtype. SOX11 is exclusively expressed in fetal and adult brain and in adult heart. Targeted disruption of Sox11 in mice resulted in a 23% birth weight reduction and lethality after the first postnatal week in homozygotes, due to hypoplastic lungs and ventricular septation defects. In addition, skeletal malformations, including of phalanges, and abdominal defects were observed. Physical and functional abnormalities in heterozygotes had not been described. Sox11 knockdown experiments in zebrafish showed microcephaly and brain abnormalities. Tsurusaki et al. (2014) commented that SOX11 is the downstream transcriptional factor of the PAX6 (607108)-BAF (603811) complex, highlighting the importance of the BAF complex and SOX11 transcriptional network in brain development.

In a patient with IDDMOH, Wakim et al. (2021) identified a de novo heterozygous missense mutation in the HMG domain of the SOX11 gene (I49N; 600898.0003). The mutation was identified by whole-exome sequencing and confirmed by Sanger sequencing. Functional studies were not performed.

In a mother and her 2 daughters with IDDMOH, Hanker et al. (2022) identified a heterozygous missense mutation in the SOX11 gene (G47S; 600898.0004). The mutation was identified by sequencing of a next-generation sequencing panel and confirmed by Sanger sequencing. Functional studies were not performed.

In 2 unrelated boys with IDDMOH, Alburaiky et al. (2022) identified de novo heterozygous missense mutations in the SOX11 gene (R100P; 600898.0005; N76D, 600898.0006).

In 2 unrelated Chinese patients with IDDMOH, Ding et al. (2022) identified de novo heterozygous mutations in the SOX11 gene (K274X; 600898.0007; Y113H, 600898.0008). The K274X mutation was predicted to result in nonsense-mediated mRNA decay. A luciferase assay using a reporter construct with a fragment of the GDF5 promoter showed that SOX11 with the Y113H mutation resulted in decreased transcriptional activity compared to wildtype.

Al-Jawahiri et al. (2022) reported heterozygous mutations in the SOX11 gene in 38 patients, including 2 sib pairs, with IDDMOH. The mutations included 25 different missense mutations, 4 truncating mutations, and 4 microdeletions. None of the mutations were present in the gnomAD database. Most of the missense mutations were located in the HMG domain. A luciferase assay using a reporter construct with the GDF5 promoter showed that SOX11 with either the A176E, Y294X or Gly384ArgfsTer14 mutation resulted in decreased transcriptional activity compared to wildtype. The transcriptional defects were more severe in the 2 protein-truncating mutations compared to the missense mutation. Methylation analysis in peripheral blood from 10 patients with IDDMOH demonstrated a hypomethylation pattern that was distinct from other BAFopathy complex epigenetic disorders.


ALLELIC VARIANTS ( 8 Selected Examples):

.0001 INTELLECTUAL DEVELOPMENTAL DISORDER WITH MICROCEPHALY AND WITHOUT OCULAR MALFORMATIONS OR HYPOGONADOTROPIC HYPOGONADISM

SOX11, TYR116CYS
  
RCV000128428

In a Japanese girl with intellectual developmental disorder with microcephaly (IDDMOH; 615866), Tsurusaki et al. (2014) identified a c.347A-G transition in the SOX11 gene, resulting in a tyr116-to-cys (Y116C) substitution. The mutation, which occurred as a de novo event, disrupted an amino acid conserved from zebrafish to human located in the HMG domain. The mutation was not identified in the 1000 Genomes Project, Exome Variant Server, or in-house databases. The patient showed dysmorphic facial features, microcephaly, growth deficiency, hypoplastic fifth toenails, and mildly impaired intellectual development. The authors described the disorder as mild Coffin-Siris syndrome.


.0002 INTELLECTUAL DEVELOPMENTAL DISORDER WITH MICROCEPHALY AND HYPOGONADOTROPIC HYPOGONADISM

SOX11, SER60PRO
  
RCV000128429

In a 17-year-old Indian girl with intellectual developmental disorder with microcephaly and hypogonadotropic hypogonadism (IDDMOH; 615866), Tsurusaki et al. (2014) identified a c.178T-C transition in the SOX11 gene, resulting in a ser60-to-pro (S60P) substitution. The mutation, which occurred as a de novo event, disrupted an amino acid conserved from zebrafish to human located in the HMG domain. The mutation was not identified in the 1000 Genomes Project, Exome Variant Server, or in-house databases. The patients showed dysmorphic facial features, microcephaly, growth deficiency, hypoplastic fifth toenails, and mildly impaired intellectual development. Ultrasonographic examination at age 16 years showed a hypoplastic uterus. No secondary sexual characteristics were recognized until menarche at age 17. The authors described the disorder as mild Coffin-Siris syndrome.


.0003 INTELLECTUAL DEVELOPMENTAL DISORDER WITH MICROCEPHALY AND WITHOUT OCULAR MALFORMATIONS OR HYPOGONADOTROPIC HYPOGONADISM

SOX11, ILE49ASN
   RCV003150873

In a Lebanese boy with intellectual developmental disorder with microcephaly and without ocular malformations or hypogonadotropic hypogonadism (IDDMOH; 615866), Wakim et al. (2021) identified heterozygosity for a c.146T-A transition in the SOX11 gene, resulting in an ile49-to-asn (I49N) substitution at a conserved residue in the HMG domain. The mutation, which was identified by whole-exome sequencing and confirmed with Sanger sequencing, was found to be de novo. The mutation was not present in the gnomAD and 1000 Genomes databases. Functional studies were not performed.


.0004 INTELLECTUAL DEVELOPMENTAL DISORDER WITH MICROCEPHALY AND OCULAR MALFORMATIONS

SOX11, GLY47SER
   RCV003150868

In a mother and her 2 daughters with intellectual developmental disorder with microcephaly and ocular malformations (IDDMOH; 615866), Hanker et al. (2022) identified heterozygosity for a c.139G-A transition (c.139G-A, NM_003108.3) in the SOX11 gene, resulting in a gly47-to-ser (G47S) substitution. The mutation, which was identified by sequencing of a next-generation sequencing panel of 8 genes and confirmed by Sanger sequencing, segregated with disease in the family. Functional studies were not performed.


.0005 INTELLECTUAL DEVELOPMENTAL DISORDER WITH MICROCEPHALY AND OCULAR MALFORMATIONS

SOX11, ARG100PRO
  
RCV000481795...

In a boy (patient 1) with intellectual developmental disorder with microcephaly and ocular malformations (IDDMOH; 615866), who also had bilateral cryptorchidism and micropenis, Alburaiky et al. (2022) identified de novo heterozygosity for a c.299G-C transversion (299G-C, NM_003108.3) in the SOX11 gene, resulting in an arg100-to-pro (R100P) substitution. The mutation was identified by trio whole-exome sequencing. Functional studies were not performed.


.0006 INTELLECTUAL DEVELOPMENTAL DISORDER WITH OCULAR MALFORMATIONS

SOX11, ASN76ASP
  
RCV001246232...

In a boy (patient 2) with intellectual developmental disorder with ocular malformations (IDDMOH; 615866), who had a head circumference on the 25th percentile at age 2 years, an underdeveloped scrotum, and palpable testes, Alburaiky et al. (2022) identified de novo heterozygosity for a c.226A-G transition (c.226A-G, NM_003108.4) in the SOX11 gene, resulting in an asn76-to-asp (N76D) substitution. The mutation was identified by whole-exome sequencing and confirmed by Sanger sequencing. Functional studies were not performed.


.0007 INTELLECTUAL DEVELOPMENTAL DISORDER WITH OCULAR MALFORMATIONS

SOX11, LYS274TER
   RCV003150872

In a Chinese girl (patient 2) with intellectual developmental disorder with ocular malformations (IDDMOH; 615866), Ding et al. (2022) identified de novo heterozygosity for a c.820A-T transversion (c.820A-T, NM_003108.4) in the SOX11 gene, resulting in a lys274-to-ter (K274X) substitution. The mutation was identified by whole-exome sequencing and confirmed by Sanger sequencing. The mutation was predicted to result in nonsense-mediated mRNA decay. (In the article by Ding et al. (2022), the mutation is given as K142X in the abstract, but as K274X in figure 2 and the text.)


.0008 INTELLECTUAL DEVELOPMENTAL DISORDER WITH MICROCEPHALY AND WITHOUT OCULAR MALFORMATIONS OR HYPOGONADOTROPIC HYPOGONADISM

SOX11, TYR113HIS
   RCV003150869

In a Chinese boy (patient 3) with intellectual developmental disorder and microcephaly (IDDMOH; 615866), Ding et al. (2022) identified a de novo heterozygous c.337T-C transition (c.337T-C, NM_003108.4) in the SOX11 gene, resulting in a tyr113-to-his (Y113H) substitution in the HMG domain. The mutation was identified by whole-exome sequencing and confirmed by Sanger sequencing. A luciferase assay using a reporter construct with a fragment of the GDF5 promoter, showed that SOX11 with the Y113H mutation resulted in decreased transcriptional activity compared to wildtype.


REFERENCES

  1. Al-Jawahiri, R., Foroutan, A., Kerkhof, J., McConkey, H., Levy, M., Haghshenas, S., Rooney, K., Turner, J., Shears, D., Holder, M., Lefroy, H., Castle, B., and 44 others. SOX11 variants cause a neurodevelopmental disorder with infrequent ocular malformations and hypogonadotropic hypogonadism and with distinct DNA methylation profile. Genet. Med. 24: 1261-1273, 2022. [PubMed: 35341651, images, related citations] [Full Text]

  2. Alburaiky, S., Taylor, J., O'Grady, G., Thomson, G., Perry, D., England, E. M., Yap, P. Cochlear nerve deficiency in SOX11-related Coffin-Siris syndrome. Am. J. Med. Genet. 188A: 2460-2465, 2022. [PubMed: 35642566, related citations] [Full Text]

  3. Ding, Y., Chen, J., Tang, Y., Chen, L. N., Yao, R. E., Yu, T., Yin, Y., Wang, X., Wang, J., Li, N. Identification and functional analysis of novel SOX11 variants in Chinese patients with Coffin-Siris syndrome 9. Front. Genet. 13: 940776, 2022. [PubMed: 35938035, images, related citations] [Full Text]

  4. Dodonova, S. O., Zhu, F., Dienemann, C., Taipale, J., Cramer, P. Nucleosome-bound SOX2 and SOX11 structures elucidate pioneer factor function. Nature 580: 669-672, 2020. [PubMed: 32350470, related citations] [Full Text]

  5. Hanker, B., Gillessen-Kaesbach, G., Huning, I., Ludecke, H. J., Wieczorek, D. Maternal transmission of a mild Coffin-Siris syndrome phenotype caused by a SOX11 missense variant. Europ. J. Hum. Genet. 30: 126-132, 2022. [PubMed: 33785884, related citations] [Full Text]

  6. Jay, P., Goze, C., Marsollier, C., Taviaux, S., Hardelin, J.-P., Koopman, P., Berta, P. The human SOX11 gene: cloning, chromosomal assignment and tissue expression. Genomics 29: 541-545, 1995. [PubMed: 8666406, related citations] [Full Text]

  7. Laudet, V., Stehelin, D., Clevers, H. Ancestry and diversity of the HMG box superfamily. Nucleic Acids Res. 21: 2493-2501, 1993. [PubMed: 8506143, related citations] [Full Text]

  8. Shim, S., Kwan, K. Y., Li, M., Lefebvre, V., Sestan, N. Cis-regulatory control of corticospinal system development and evolution. Nature 486: 74-79, 2012. [PubMed: 22678282, images, related citations] [Full Text]

  9. Tsurusaki, Y., Koshimizu, E., Ohashi, H., Phadke, S., Kou, I., Shiina, M., Suzuki, T., Okamoto, N., Imamura, S., Yamashita, M., Watanabe, S., Yoshiura, K., Kodera, H., Miyatake, S., Nakashima, M., Saitsu, H., Ogata, K., Ikegawa, S., Miyake, N., Matsumoto, N. De novo SOX11 mutations cause Coffin-Siris syndrome. Nature Commun. 5: 4011, 2014. Note: Electronic Article. [PubMed: 24886874, related citations] [Full Text]

  10. Wakim, V., Nair, P., Delague, V., Bizzari, S., Al-Ali, M. T., Castro, C., Gambarini, A., El-Hayek, S., Megarbane, A. SOX11-related syndrome: report on a new case and review. Clin. Dysmorph. 30: 44-49, 2021. [PubMed: 33086258, related citations] [Full Text]


Hilary J. Vernon - updated : 02/02/2023
Ada Hamosh - updated : 06/08/2020
Ada Hamosh - updated : 6/26/2014
Ada Hamosh - updated : 7/17/2012
Creation Date:
Victor A. McKusick : 10/25/1995
carol : 03/14/2023
carol : 02/06/2023
carol : 02/03/2023
carol : 02/02/2023
carol : 02/02/2023
alopez : 06/08/2020
alopez : 07/16/2019
carol : 11/06/2014
alopez : 6/26/2014
alopez : 7/17/2012
terry : 7/17/2012
ckniffin : 10/15/2003
dkim : 12/4/1998
carol : 5/27/1998
mark : 6/25/1996
mark : 10/25/1995

* 600898

SRY-BOX 11; SOX11


Alternative titles; symbols

SRY-RELATED HMG-BOX GENE 11


HGNC Approved Gene Symbol: SOX11

Cytogenetic location: 2p25.2     Genomic coordinates (GRCh38): 2:5,692,384-5,701,385 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
2p25.2 Intellectual developmental disorder with microcephaly and with or without ocular malformations or hypogonadotropic hypogonadism 615866 Autosomal dominant 3

TEXT

Cloning and Expression

Using the partial clones of both human and mouse SOX11 genes, Jay et al. (1995) cloned and characterized the human SOX11 gene. The SOX11 sequence is strongly conserved with the chicken homolog and is related to SOX4. It contains several putative transcriptional activator or repressor domains. The authors observed that the SOX11 expression pattern is consistent with the hypothesis that this gene is important in the developing nervous system.


Mapping

Jay et al. (1995) mapped the SOX11 gene to chromosome 2p25 by fluorescence in situ hybridization.


Gene Function

Shim et al. (2012) identified a conserved nonexonic element (E4), located 7.3 kb downstream of the Fezf2 (607414) transcription start site, that is required for the specification of corticospinal neuron identity and connectivity. Shim et al. (2012) found that Sox4 (184430) and Sox11 functionally compete with the repressor Sox5 (604975) in the transactivation of E4. Shim et al. (2012) showed that SOX4 and SOX11 are crucial in regulating reelin (RELN; 600514) expression and the inside-out pattern of cortical layer formation, independent of E4 or Fezf2 and probably involving interactions with distinct regulatory elements. Cortex-specific double deletion of Sox4 and Sox11 led to the loss of Fezf2 expression, failed specification of corticospinal neurons and, independent of Fezf2, a reeler-like inversion of layers. Moreover, SOX4 and SOX11 have additional roles, since in mice lacking both genes, the cortex and olfactory bulb are smaller and cell death is increased. Thus, SOX4 and SOX11 have pleiotropic functions, which are probably mediated by distinct regulatory elements and downstream target genes that are involved in multiple developmental processes. Shim et al. (2012) showed evidence supporting the emergence of functional SOX-binding sites in E4 during tetrapod evolution, and their subsequent stabilization in mammals and possibly amniotes. Shim et al. (2012) concluded that SOX transcription factors converge onto a cis-acting element of Fezf2 and form critical components of a regulatory network controlling the identity and connectivity of corticospinal neurons.


Biochemical Features

Cryoelectron Microscopy

Dodonova et al. (2020) reported cryoelectron microscopy structures of the DNA-binding domains of SOX2 (184429) and its close homolog SOX11 bound to nucleosomes. The structures showed that SOX factors can bind and locally distort DNA at superhelical location 2. The factors also facilitated detachment of terminal nucleosomal DNA from the histone octamer, which increases DNA accessibility. SOX-factor binding to the nucleosome can also lead to a repositioning of the N-terminal tail of histone H4 (see 602822) that includes residue lys16. Dodonova et al. (2020) speculated that this repositioning is incompatible with higher-order nucleosome stacking, which involves contacts of the H4 tail with a neighboring nucleosome. Dodonova et al. (2020) concluded that pioneer transcription factors that maintain pluripotency can use binding energy to initiate chromatin opening, and thereby facilitate nucleosome remodeling and subsequent transcription.


Gene Family

SRY (480000) is the testis-determining gene located on the Y chromosome of mammals. It encodes a protein whose most striking feature is a motif of 78 amino acids conserved with respect to the DNA binding domain of the high mobility group (HMG) proteins. Jay et al. (1995) noted that more than 100 HMG box-containing proteins had been reported at that time and are classified in 2 distinct subgroups according to the sequence-specificity of the binding, the number of DNA binding domains, and phylogenetic considerations (Laudet et al., 1993). An important subgroup of HMG box-containing proteins includes SRY and SRY box-related (SOX) sequences. They contain only 1 DNA-binding domain, and they bind to DNA in a sequence-specific manner. They are all potential transcription factors implicated in the developmental control of gene expression. Degenerate PCR-based methods enabled the cloning and sequencing of a great number of new SRY-related box sequences in both vertebrates and invertebrates.


Molecular Genetics

Tsurusaki et al. (2014) identified 2 de novo missense mutations in the SOX11 gene (Y116C, 600898.0001 and S60P, 600898.0002) in 2 unrelated female patients with intellectual developmental disorder with microcephaly and with or without ocular malformations or hypogonadotropic hypogonadism (IDDMOH; 615866), also referred to as Coffin-Siris syndrome-9 (CSS9). Both mutations occurred in the HMG domain in 2 evolutionarily conserved amino acids. Tsurusaki et al. (2014) showed that both mutations caused decreased transcriptional activation compared to wildtype. SOX11 is exclusively expressed in fetal and adult brain and in adult heart. Targeted disruption of Sox11 in mice resulted in a 23% birth weight reduction and lethality after the first postnatal week in homozygotes, due to hypoplastic lungs and ventricular septation defects. In addition, skeletal malformations, including of phalanges, and abdominal defects were observed. Physical and functional abnormalities in heterozygotes had not been described. Sox11 knockdown experiments in zebrafish showed microcephaly and brain abnormalities. Tsurusaki et al. (2014) commented that SOX11 is the downstream transcriptional factor of the PAX6 (607108)-BAF (603811) complex, highlighting the importance of the BAF complex and SOX11 transcriptional network in brain development.

In a patient with IDDMOH, Wakim et al. (2021) identified a de novo heterozygous missense mutation in the HMG domain of the SOX11 gene (I49N; 600898.0003). The mutation was identified by whole-exome sequencing and confirmed by Sanger sequencing. Functional studies were not performed.

In a mother and her 2 daughters with IDDMOH, Hanker et al. (2022) identified a heterozygous missense mutation in the SOX11 gene (G47S; 600898.0004). The mutation was identified by sequencing of a next-generation sequencing panel and confirmed by Sanger sequencing. Functional studies were not performed.

In 2 unrelated boys with IDDMOH, Alburaiky et al. (2022) identified de novo heterozygous missense mutations in the SOX11 gene (R100P; 600898.0005; N76D, 600898.0006).

In 2 unrelated Chinese patients with IDDMOH, Ding et al. (2022) identified de novo heterozygous mutations in the SOX11 gene (K274X; 600898.0007; Y113H, 600898.0008). The K274X mutation was predicted to result in nonsense-mediated mRNA decay. A luciferase assay using a reporter construct with a fragment of the GDF5 promoter showed that SOX11 with the Y113H mutation resulted in decreased transcriptional activity compared to wildtype.

Al-Jawahiri et al. (2022) reported heterozygous mutations in the SOX11 gene in 38 patients, including 2 sib pairs, with IDDMOH. The mutations included 25 different missense mutations, 4 truncating mutations, and 4 microdeletions. None of the mutations were present in the gnomAD database. Most of the missense mutations were located in the HMG domain. A luciferase assay using a reporter construct with the GDF5 promoter showed that SOX11 with either the A176E, Y294X or Gly384ArgfsTer14 mutation resulted in decreased transcriptional activity compared to wildtype. The transcriptional defects were more severe in the 2 protein-truncating mutations compared to the missense mutation. Methylation analysis in peripheral blood from 10 patients with IDDMOH demonstrated a hypomethylation pattern that was distinct from other BAFopathy complex epigenetic disorders.


ALLELIC VARIANTS 8 Selected Examples):

.0001   INTELLECTUAL DEVELOPMENTAL DISORDER WITH MICROCEPHALY AND WITHOUT OCULAR MALFORMATIONS OR HYPOGONADOTROPIC HYPOGONADISM

SOX11, TYR116CYS
SNP: rs587777479, ClinVar: RCV000128428

In a Japanese girl with intellectual developmental disorder with microcephaly (IDDMOH; 615866), Tsurusaki et al. (2014) identified a c.347A-G transition in the SOX11 gene, resulting in a tyr116-to-cys (Y116C) substitution. The mutation, which occurred as a de novo event, disrupted an amino acid conserved from zebrafish to human located in the HMG domain. The mutation was not identified in the 1000 Genomes Project, Exome Variant Server, or in-house databases. The patient showed dysmorphic facial features, microcephaly, growth deficiency, hypoplastic fifth toenails, and mildly impaired intellectual development. The authors described the disorder as mild Coffin-Siris syndrome.


.0002   INTELLECTUAL DEVELOPMENTAL DISORDER WITH MICROCEPHALY AND HYPOGONADOTROPIC HYPOGONADISM

SOX11, SER60PRO
SNP: rs587777480, ClinVar: RCV000128429

In a 17-year-old Indian girl with intellectual developmental disorder with microcephaly and hypogonadotropic hypogonadism (IDDMOH; 615866), Tsurusaki et al. (2014) identified a c.178T-C transition in the SOX11 gene, resulting in a ser60-to-pro (S60P) substitution. The mutation, which occurred as a de novo event, disrupted an amino acid conserved from zebrafish to human located in the HMG domain. The mutation was not identified in the 1000 Genomes Project, Exome Variant Server, or in-house databases. The patients showed dysmorphic facial features, microcephaly, growth deficiency, hypoplastic fifth toenails, and mildly impaired intellectual development. Ultrasonographic examination at age 16 years showed a hypoplastic uterus. No secondary sexual characteristics were recognized until menarche at age 17. The authors described the disorder as mild Coffin-Siris syndrome.


.0003   INTELLECTUAL DEVELOPMENTAL DISORDER WITH MICROCEPHALY AND WITHOUT OCULAR MALFORMATIONS OR HYPOGONADOTROPIC HYPOGONADISM

SOX11, ILE49ASN
ClinVar: RCV003150873

In a Lebanese boy with intellectual developmental disorder with microcephaly and without ocular malformations or hypogonadotropic hypogonadism (IDDMOH; 615866), Wakim et al. (2021) identified heterozygosity for a c.146T-A transition in the SOX11 gene, resulting in an ile49-to-asn (I49N) substitution at a conserved residue in the HMG domain. The mutation, which was identified by whole-exome sequencing and confirmed with Sanger sequencing, was found to be de novo. The mutation was not present in the gnomAD and 1000 Genomes databases. Functional studies were not performed.


.0004   INTELLECTUAL DEVELOPMENTAL DISORDER WITH MICROCEPHALY AND OCULAR MALFORMATIONS

SOX11, GLY47SER
ClinVar: RCV003150868

In a mother and her 2 daughters with intellectual developmental disorder with microcephaly and ocular malformations (IDDMOH; 615866), Hanker et al. (2022) identified heterozygosity for a c.139G-A transition (c.139G-A, NM_003108.3) in the SOX11 gene, resulting in a gly47-to-ser (G47S) substitution. The mutation, which was identified by sequencing of a next-generation sequencing panel of 8 genes and confirmed by Sanger sequencing, segregated with disease in the family. Functional studies were not performed.


.0005   INTELLECTUAL DEVELOPMENTAL DISORDER WITH MICROCEPHALY AND OCULAR MALFORMATIONS

SOX11, ARG100PRO
SNP: rs1064794628, gnomAD: rs1064794628, ClinVar: RCV000481795, RCV003150820

In a boy (patient 1) with intellectual developmental disorder with microcephaly and ocular malformations (IDDMOH; 615866), who also had bilateral cryptorchidism and micropenis, Alburaiky et al. (2022) identified de novo heterozygosity for a c.299G-C transversion (299G-C, NM_003108.3) in the SOX11 gene, resulting in an arg100-to-pro (R100P) substitution. The mutation was identified by trio whole-exome sequencing. Functional studies were not performed.


.0006   INTELLECTUAL DEVELOPMENTAL DISORDER WITH OCULAR MALFORMATIONS

SOX11, ASN76ASP
SNP: rs1665660543, ClinVar: RCV001246232, RCV003150825

In a boy (patient 2) with intellectual developmental disorder with ocular malformations (IDDMOH; 615866), who had a head circumference on the 25th percentile at age 2 years, an underdeveloped scrotum, and palpable testes, Alburaiky et al. (2022) identified de novo heterozygosity for a c.226A-G transition (c.226A-G, NM_003108.4) in the SOX11 gene, resulting in an asn76-to-asp (N76D) substitution. The mutation was identified by whole-exome sequencing and confirmed by Sanger sequencing. Functional studies were not performed.


.0007   INTELLECTUAL DEVELOPMENTAL DISORDER WITH OCULAR MALFORMATIONS

SOX11, LYS274TER
ClinVar: RCV003150872

In a Chinese girl (patient 2) with intellectual developmental disorder with ocular malformations (IDDMOH; 615866), Ding et al. (2022) identified de novo heterozygosity for a c.820A-T transversion (c.820A-T, NM_003108.4) in the SOX11 gene, resulting in a lys274-to-ter (K274X) substitution. The mutation was identified by whole-exome sequencing and confirmed by Sanger sequencing. The mutation was predicted to result in nonsense-mediated mRNA decay. (In the article by Ding et al. (2022), the mutation is given as K142X in the abstract, but as K274X in figure 2 and the text.)


.0008   INTELLECTUAL DEVELOPMENTAL DISORDER WITH MICROCEPHALY AND WITHOUT OCULAR MALFORMATIONS OR HYPOGONADOTROPIC HYPOGONADISM

SOX11, TYR113HIS
ClinVar: RCV003150869

In a Chinese boy (patient 3) with intellectual developmental disorder and microcephaly (IDDMOH; 615866), Ding et al. (2022) identified a de novo heterozygous c.337T-C transition (c.337T-C, NM_003108.4) in the SOX11 gene, resulting in a tyr113-to-his (Y113H) substitution in the HMG domain. The mutation was identified by whole-exome sequencing and confirmed by Sanger sequencing. A luciferase assay using a reporter construct with a fragment of the GDF5 promoter, showed that SOX11 with the Y113H mutation resulted in decreased transcriptional activity compared to wildtype.


REFERENCES

  1. Al-Jawahiri, R., Foroutan, A., Kerkhof, J., McConkey, H., Levy, M., Haghshenas, S., Rooney, K., Turner, J., Shears, D., Holder, M., Lefroy, H., Castle, B., and 44 others. SOX11 variants cause a neurodevelopmental disorder with infrequent ocular malformations and hypogonadotropic hypogonadism and with distinct DNA methylation profile. Genet. Med. 24: 1261-1273, 2022. [PubMed: 35341651] [Full Text: https://doi.org/10.1016/j.gim.2022.02.013]

  2. Alburaiky, S., Taylor, J., O'Grady, G., Thomson, G., Perry, D., England, E. M., Yap, P. Cochlear nerve deficiency in SOX11-related Coffin-Siris syndrome. Am. J. Med. Genet. 188A: 2460-2465, 2022. [PubMed: 35642566] [Full Text: https://doi.org/10.1002/ajmg.a.62851]

  3. Ding, Y., Chen, J., Tang, Y., Chen, L. N., Yao, R. E., Yu, T., Yin, Y., Wang, X., Wang, J., Li, N. Identification and functional analysis of novel SOX11 variants in Chinese patients with Coffin-Siris syndrome 9. Front. Genet. 13: 940776, 2022. [PubMed: 35938035] [Full Text: https://doi.org/10.3389/fgene.2022.940776]

  4. Dodonova, S. O., Zhu, F., Dienemann, C., Taipale, J., Cramer, P. Nucleosome-bound SOX2 and SOX11 structures elucidate pioneer factor function. Nature 580: 669-672, 2020. [PubMed: 32350470] [Full Text: https://doi.org/10.1038/s41586-020-2195-y]

  5. Hanker, B., Gillessen-Kaesbach, G., Huning, I., Ludecke, H. J., Wieczorek, D. Maternal transmission of a mild Coffin-Siris syndrome phenotype caused by a SOX11 missense variant. Europ. J. Hum. Genet. 30: 126-132, 2022. [PubMed: 33785884] [Full Text: https://doi.org/10.1038/s41431-021-00865-2]

  6. Jay, P., Goze, C., Marsollier, C., Taviaux, S., Hardelin, J.-P., Koopman, P., Berta, P. The human SOX11 gene: cloning, chromosomal assignment and tissue expression. Genomics 29: 541-545, 1995. [PubMed: 8666406] [Full Text: https://doi.org/10.1006/geno.1995.9970]

  7. Laudet, V., Stehelin, D., Clevers, H. Ancestry and diversity of the HMG box superfamily. Nucleic Acids Res. 21: 2493-2501, 1993. [PubMed: 8506143] [Full Text: https://doi.org/10.1093/nar/21.10.2493]

  8. Shim, S., Kwan, K. Y., Li, M., Lefebvre, V., Sestan, N. Cis-regulatory control of corticospinal system development and evolution. Nature 486: 74-79, 2012. [PubMed: 22678282] [Full Text: https://doi.org/10.1038/nature11094]

  9. Tsurusaki, Y., Koshimizu, E., Ohashi, H., Phadke, S., Kou, I., Shiina, M., Suzuki, T., Okamoto, N., Imamura, S., Yamashita, M., Watanabe, S., Yoshiura, K., Kodera, H., Miyatake, S., Nakashima, M., Saitsu, H., Ogata, K., Ikegawa, S., Miyake, N., Matsumoto, N. De novo SOX11 mutations cause Coffin-Siris syndrome. Nature Commun. 5: 4011, 2014. Note: Electronic Article. [PubMed: 24886874] [Full Text: https://doi.org/10.1038/ncomms5011]

  10. Wakim, V., Nair, P., Delague, V., Bizzari, S., Al-Ali, M. T., Castro, C., Gambarini, A., El-Hayek, S., Megarbane, A. SOX11-related syndrome: report on a new case and review. Clin. Dysmorph. 30: 44-49, 2021. [PubMed: 33086258] [Full Text: https://doi.org/10.1097/MCD.0000000000000348]


Contributors:
Hilary J. Vernon - updated : 02/02/2023
Ada Hamosh - updated : 06/08/2020
Ada Hamosh - updated : 6/26/2014
Ada Hamosh - updated : 7/17/2012

Creation Date:
Victor A. McKusick : 10/25/1995

Edit History:
carol : 03/14/2023
carol : 02/06/2023
carol : 02/03/2023
carol : 02/02/2023
carol : 02/02/2023
alopez : 06/08/2020
alopez : 07/16/2019
carol : 11/06/2014
alopez : 6/26/2014
alopez : 7/17/2012
terry : 7/17/2012
ckniffin : 10/15/2003
dkim : 12/4/1998
carol : 5/27/1998
mark : 6/25/1996
mark : 10/25/1995