Entry - *300040 - STRUCTURAL MAINTENANCE OF CHROMOSOMES 1A; SMC1A - OMIM
* 300040

STRUCTURAL MAINTENANCE OF CHROMOSOMES 1A; SMC1A


Alternative titles; symbols

SMC1-ALPHA
STRUCTURAL MAINTENANCE OF CHROMOSOMES 1-LIKE 1; SMC1L1
SMC1
DXS423E
KIAA0178


HGNC Approved Gene Symbol: SMC1A

Cytogenetic location: Xp11.22     Genomic coordinates (GRCh38): X:53,374,149-53,422,728 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
Xp11.22 Cornelia de Lange syndrome 2 300590 XLD 3
Developmental and epileptic encephalopathy 85, with or without midline brain defects 301044 XLD 3

TEXT

Description

Eukaryotic sister chromatids remain connected from the time of synthesis until they are separated in anaphase. This cohesion depends on a complex of proteins known as cohesins. In vertebrates, unlike in yeast, the cohesins dissociate from chromosome arms earlier in M phase, during prophase. Small amounts of cohesin remain near the centromere until metaphase, with complete removal at the beginning of anaphase. Cohesin complexes contain SMC1, SCC1 (RAD21), SMC3 (606062), and either SA1 (STAG1; 604358) or SA2 (STAG2; 300826). The complexes, in turn, interact with PDS5 (see 613200), a protein implicated in chromosome cohesion, condensation, and recombination in yeast (summary by Sumara et al., 2000).


Cloning and Expression

Genetic marker SB1.8 (DXS423E) was originally identified as a cross-reacting clone during the screening of a human lymphocyte cDNA library with an oligonucleotide probe corresponding to the CYBB (300481) gene, which maps to Xp21.1. Rocques et al. (1995) found that the DXS423E gene encodes a protein of 1,233 amino acids that is 30% identical to the essential yeast protein SMC1 (structural maintenance of chromosomes-1), which is required for the segregation of chromosomes at mitosis. Both the human protein, called SB1.8, and SMC1 contain an N-terminal NTP-binding site, a central coiled-coil region, and a C-terminal helix-loop-helix domain, and both have structural features in common with the force-generating proteins myosin and kinesin. SB1.8 also exhibits regions of homology and overall structural similarity to protein 115p of the prokaryote Mycoplasma hyorhinis.

In mouse embryos, Kruszka et al. (2019) found expression of the Smc1a gene in anterior neural folds, the neuroectoderm, and adjacent mesenchyme of the developing brain. The findings suggested a role in forebrain patterning.


Gene Function

In yeast, the cohesin complex is essential for sister chromatid cohesion during mitosis. The Smc1 and Smc3 subunits are rod-shaped molecules with globular ABC-like ATPases at one end and dimerization domains at the other, connected by long coiled coils. Smc1 and Smc3 associate to form V-shaped heterodimers. Their ATPase heads are thought to be bridged by a third subunit, Scc1, creating a huge triangular ring that can trap sister DNA molecules. Gruber et al. (2003) studied whether cohesin forms such rings in vivo. Proteolytic cleavage of Scc1 by separase at the onset of anaphase triggers its dissociation from chromosomes. The authors showed that N- and C-terminal Scc1 cleavage fragments remain connected due to their association with different heads of a single Smc1/Smc3 heterodimer. Cleavage of the Smc3 coiled coil was sufficient to trigger cohesin release from chromosomes and loss of sister cohesion, consistent with a topologic association with chromatin.

By yeast 2-hybrid screening of a human fetal brain expression cDNA library using the hinge domain of SMC1 as bait, followed by immunoprecipitation analysis, Patel and Ghiselli (2005) found that SMC1 interacted with hinderin (KIAA1328; 616480). Hinderin did not interact with SMC3. Interaction of hinderin with SMC1 precluded dimerization of SMC1 and SMC3.

Musio et al. (2005) demonstrated that RNA interference (RNAi) of SMC1 was sufficient to induce fragile site expression in normal human fibroblasts. They showed that aphidicolin treatment led to an increase in SMC1 synthesis, SMC1 phosphorylation via an ATR (601215)-dependent pathway, and enhanced double-stranded break induction as visualized by immunohistochemical studies with phosphorylated H2AX (601772). Discrete nuclear foci were absent or very rare after 1 or 2 hours exposure to aphidicolin and/or RNAi of SMC1 but became more numerous and distinct after 6 hours. Musio et al. (2005) proposed that fragile sites might be viewed as an in vitro phenomenon originating from double-strand breaks formed because of stalled DNA replication that lasted too long to be rescued via the ATR/SMC1 axis, whereas in vivo, following an extreme replication block, rare cells could escape checkpoint mechanisms and enter mitosis with a defect in genome assembly, eventually leading to neoplastic transformation.

Cohesin's Scc1, Smc1, and Smc3 subunits form a tripartite ring structure, and it had been proposed that cohesin holds sister DNA molecules together by trapping them inside its ring. To test this, Haering et al. (2008) used site-specific crosslinking to create chemical connections at the 3 interfaces between the 3 constituent polypeptides of the ring, thereby creating covalently closed cohesin rings. As predicted by the ring entrapment model, this procedure produced dimeric DNA-cohesin structures that are resistant to protein denaturation. Haering et al. (2008) concluded that cohesin rings concatenate individual sister minichromosome DNA molecules.

Kagey et al. (2010) reported that Mediator (see MED8, 607956) and cohesin physically and functionally connect the enhancers and core promoters of active genes in murine embryonic stem cells. Mediator, a transcriptional coactivator, forms a complex with cohesin, which can form rings that connect 2 DNA segments. The cohesin-loading factor NIPBL (608667) is associated with Mediator-cohesin complexes, providing a means to load cohesin at promoters. DNA looping is observed between the enhancers and promoters occupied by Mediator and cohesin. Mediator and cohesin co-occupy different promoters in different cells, thus generating cell type-specific DNA loops linked to the gene expression program of each cell.

Using biochemical reconstitution, Davidson et al. (2019) found that single human cohesin complexes form DNA loops symmetrically at rates up to 2.1 kilobase pairs per second. Loop formation and maintenance depend on cohesin's ATPase activity and on NIPBL-MAU2 (614560), but not on topologic entrapment of DNA by cohesin (components include SMC3, 606062; SMC1A; STAG1, 604358; and STAG2, 300826). During loop formation, cohesin and NIPBL-MAU2 reside at the base of loops, which indicates that they generate loops by extrusion. Davidson et al. (2019) concluded that their results showed that cohesin and NIPBL-MAU2 form an active holoenzyme that interacts with DNA either pseudotopologically or nontopologically to extrude genomic interphase DNA into loops.


Mapping

Rocques et al. (1995) showed that the DXS423E gene maps to a cosmid contig that lies centromeric to the OATL2 locus (see 258870) at Xp11.2. Brown et al. (1995) showed that SMC1 escapes X-chromosome inactivation. SMC1 and XE169 (314690) were thought to define a new region in the proximal short arm of the X chromosome that escapes X inactivation. The corresponding gene in the mouse, Sb1.8, is located at the distal end of the X chromosome and is subject to X inactivation (Sultana et al., 1995).


Molecular Genetics

Cornelia de Lange Syndrome 2

A female with a Cornelia de Lange syndrome (122470) phenotype carrying an apparently balanced X;8 translocation involving the Xp11.2 band, to which the SMC1L1 gene maps, was described by Egemen et al. (2005). The findings were consistent with the location of the SMC1L1 gene at Xp11.2 and the involvement of that gene in X-linked Cornelia de Lange syndrome-2 (CDLS2; 300590).

Musio et al. (2006) recruited 53 unrelated and 4 related individuals with a diagnosis of Cornelia de Lange syndrome, encompassing the entire spectrum of phenotypes. They found pathogenic NIPBL (608667) mutations in 24 of them, whereas the remaining 33 cases did not bear any NIPBL mutation. Of these 33 individuals, there was only 1 instance of familial occurrence, with 2 male sibs, their mother, and a first cousin affected. Involvement of the NIPBL gene was excluded in this family, but the affected individuals were found to carry a 3-bp deletion in the SMC1L1 gene (300040.0001). In addition, a sporadic case was found to have a de novo missense mutation in the SMC1L1 gene (300040.0002).

Deardorff et al. (2007) identified 14 additional SMC1A mutations in patients with a mild variant of Cornelia de Lange syndrome with predominant mental retardation. Analysis of the mutant SMC1A proteins indicated that they were likely to produce functional cohesin complexes; however, Deardorff et al. (2007) posited that they mutations may alter their chromosome binding dynamics. Ten of 14 SMC1A-mutation-positive individuals with CDLS identified by Deardorff et al. (2007) were female. Furthermore, their series included similarly affected male and female probands, implying an X-linked dominant mode of expression. Several males were rather mildly affected and no more severely affected than many of the SMC1A mutation-positive females. Since the SMC1A gene escapes X inactivation (Brown et al., 1995), it is likely that the mechanism in affected females is due to a dominant-negative effect of the altered protein and less likely that it is due to decreased protein levels or skewed X inactivation. Consistent with this dominant-negative effect on cohesin, Deardorff et al. (2007) described a single amino acid deletion mutation in the SMC3 gene underlying a variant Cornelia de Lange syndrome (606062.0001). The data indicated that SMC3 and SMC1A mutations contribute to approximately 5% of cases of Cornelia de Lange syndrome, result in a consistently mild phenotype with absence of major structural anomalies typically associated with CDLS, such as those of the limbs, and, in some instances, result in a phenotype that approaches that of apparently nonsyndromic mental retardation. Deardorff et al. (2007) suggested that it may be found that additional 'cohesinopathies' result from perturbation of the more than 15 additional components of this complex that had yet to be associated with human disorders.

Developmental and Epileptic Encephalopathy 85 with or without Midline Brain Defects

In a 7-year-old girl, born of unrelated Portuguese parents, with developmental and epileptic encephalopathy-85 with midline brain defects manifest as thin corpus callosum (DEE85; 301044), Lebrun et al. (2015) identified a de novo heterozygous splice site mutation in the SMC1A gene (300040.0007). The mutation was found by exome sequencing and confirmed by Sanger sequencing. Analysis of patient fibroblasts showed the presence of only the mutant transcript, which was significantly reduced compared to controls, suggesting nonsense-mediated mRNA decay and a loss of function.

In 2 unrelated girls with DEE85, Goldstein et al. (2015) identified de novo heterozygous frameshift mutations in the SMC1A gene (300040.0008 and 300040.0009). The mutations were found by whole-exome sequencing and confirmed by Sanger sequencing. X-inactivation studies in peripheral blood cells showed a skewed pattern (93:7) in 1 patient, but a random pattern in the other. Additional functional studies of the variants and studies of patient cells were not performed.

In 2 unrelated females with DEE85, Jansen et al. (2016) identified de novo heterozygous loss of function (LOF) mutations in the SMC1A gene (see, e.g., 300040.0010). The mutations were found by whole-genome sequencing. X-inactivation studies in 1 patient showed a random pattern, whereas it was skewed in the other patient (85:15). Additional functional studies were not performed. Jansen et al. (2016) concluded that the de novo LOF mutations in the SMC1A gene in females cause an abnormal dosage effect and that the resulting phenotype is distinct from CDLS2. The authors further suggested that LOF mutations in males may be embryonic lethal.

In 10 unrelated females with DEE85, Symonds et al. (2017) identified de novo heterozygous nonsense, frameshift, or splice site mutations in the SMC1A gene (see, e.g., 300040.0011-300040.0013). Functional studies of the variants were not performed. X-inactivation studies, performed in some patients, had variable results: some showed a random pattern, whereas others had a skewed pattern. Symonds et al. (2017) speculated that if SMC1A escapes X inactivation, then haploinsufficiency is unlikely to be the causative mechanism; rather, the authors postulated a dominant-negative effect. The findings also suggested that complete SMC1A deficiency is embryonic lethal, as no males with such mutations have been reported.

In 5 unrelated girls (patients 7-11) with DEE85 and variable midline brain defects, including holoprosencephaly (HPE), Kruszka et al. (2019) identified de novo heterozygous mutations in the SMC1A gene (see, e.g., 300040.0014 and 300040.0015). All mutations except 1 were nonsense, frameshift, or splice site mutations, suggesting a loss-of-function effect; there was 1 missense mutation. Functional studies of the variants and studies of patient cells were not performed. Knockdown of the SMC1A gene in human neural progenitor cells resulted in upregulation of GLI2 (165230), ZIC2 (603073), and SMAD3 (603109) gene expression. Although the significance of these findings was unclear, it demonstrated that loss of SMC1A perturbs the expression of genes involved in HPE. The patients were part of a cohort of 277 individuals with HPE as well as gathered through collaborative efforts such as GeneMatcher.


ALLELIC VARIANTS ( 15 Selected Examples):

.0001 CORNELIA DE LANGE SYNDROME 2

SMC1A, 3-BP DEL, 2493CCA
  
RCV000012438

Musio et al. (2006) identified a 3-bp deletion in the SMC1L1 gene in hemizygous state in 2 brothers and a maternal first cousin from a family with X-linked Cornelia de Lange syndrome (300590). The mother of the 2 brothers was heterozygous for the mutation and mildly affected. The deletion involved the third nucleotide of codon 831 and the first 2 nucleotides of codon 832, leading to deletion of gln832 and an asp831-to-glu (D831E) substitution. The mutation was not identified in over 600 control chromosomes.


.0002 CORNELIA DE LANGE SYNDROME 2

SMC1A, GLU493ALA
  
RCV000012439

In a male patient with sporadic X-linked Cornelia de Lange syndrome (300590), Musio et al. (2006) identified hemizygosity for a 1478A-C transversion in the SMC1L1 gene, resulting in an glu493-to-ala (E493A) substitution. The mutation was not identified in over 400 control chromosomes.

Revenkova et al. (2009) showed that E493A-mutant SMC1A affected the affinity of SMC hinge dimers for DNA. Mutated hinge dimers bound DNA with higher affinity than wildtype proteins. SMC1A-mutated Cornelia de Lange syndrome cell lines displayed genomic instability and sensitivity to ionizing radiation and interstrand crosslinking agents.


.0003 CORNELIA DE LANGE SYNDROME 2

SMC1A, 15-BP DEL, NT173
   RCV000012440

In a boy with a milder variant of Cornelia de Lange syndrome (CDLS2; 300590), Deardorff et al. (2007) identified a 15-bp deletion in the SMC1A gene, resulting in deletion of 5 amino acids (V58_R62del). Features of CDLS included arched eyebrows and synophrys, anteverted nostrils, long and featureless philtrum, thin lips, downturned corners of the mouth, hearing loss, cutis marmorata, small hands and feet, proximally set thumbs, clinodactyly of fifth finger, and hirsutism. The child had psychomotor delay but was in mainstream second grade schooling. He had mild pulmonic stenosis and gastroesophageal reflux.


.0004 CORNELIA DE LANGE SYNDROME 2

SMC1A, ARG496HIS
  
RCV000012441...

In 2 sisters with a variant form of Cornelia de Lange syndrome (CDLS2; 300590), Deardorff et al. (2007) identified a 1487G-A transition in the SMC1A gene that resulted in an arg496-to-his substitution (R496H). Both had psychomotor delay and moderately severe mental retardation. One had pulmonic stenosis. Both had gastroesophageal reflux.

Revenkova et al. (2009) showed that R496H-mutant SMC1A affected the affinity of SMC hinge dimers for DNA. Mutated hinge dimers bound DNA with higher affinity than wildtype proteins. SMC1A-mutated Cornelia de Lange syndrome cell lines displayed genomic instability and sensitivity to ionizing radiation and interstrand crosslinking agents.


.0005 CORNELIA DE LANGE SYNDROME 2

SMC1A, ILE784THR
  
RCV000022820...

In a 6-year-old girl with Cornelia de Lange syndrome (CDLS2; 300590), Limongelli et al. (2010) identified a de novo heterozygous 2351T-C transition in exon 15 of the SMC1A gene, resulting in an ile784-to-thr (I784T) substitution in a highly conserved residue in the coiled-coil domain. She had pre- and postnatal growth retardation, developmental delay, characteristic facial features, and concentric left ventricular hypertrophic cardiomyopathy.


.0006 CORNELIA DE LANGE SYNDROME 2

SMC1A, 8.152-KB DEL
   RCV000022821

In a girl with a severe form of Cornelia de Lange syndrome-2 (CDLS2; 300590), Hoppman-Chaney et al. (2012) identified a de novo heterozygous 8.152-kb deletion encompassing exon 13 to intron 16 of the SMC1A gene, resulting in an in-frame deletion of 126 amino acids and insertion of 3 novel amino acids. The mutant mRNA was expressed, and Hoppman-Chaney et al. (2012) concluded that the mutant protein lacking the coiled-coil domain would act in a dominant-negative manner. The patient had dysmorphic facial features, microcephaly, poor growth, profound psychomotor retardation, holoprosencephaly, right hemihypertrophy, and mild distal limb anomalies. The patient was also mosaic for Turner syndrom, 45,X(7)/46,XX(23), which may have accounted for some additional features.


.0007 DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 85 WITH MIDLINE BRAIN DEFECTS

SMC1A, IVS11DS, G-T, +1
  
RCV001072123

In a 7-year-old girl, born of unrelated Portuguese parents, with developmental and epileptic encephalopathy-85 and thin corpus callosum (DEE85; 301044), Lebrun et al. (2015) identified a de novo heterozygous G-to-T transversion (c.1911+1G-T) in intron 11 of the SMC1A gene, predicted to result in a splicing defect, frameshift, and premature termination (Thr638ValfsTer48). The mutation was found by exome sequencing and confirmed by Sanger sequencing. Analysis of patient fibroblasts showed the presence of only the mutant transcript, which was significantly reduced compared to controls, suggesting nonsense-mediated mRNA decay and a loss of function. The patient had onset of infantile spasms associated with hypsarrhythmia on EEG within the first month of life. She also had microcephaly, dysmorphic facial features, and severe developmental delay. Brain imaging showed a thin corpus callosum.


.0008 DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 85 WITH MIDLINE BRAIN DEFECTS

SMC1A, 4-BP DEL, 2853TCAG
  
RCV000202429...

In a 4-year-old girl (patient A) with developmental and epileptic encephalopathy-85 (DEE85; 301044) with possible thickening of the insular cortex on brain imaging, Goldstein et al. (2015) identified a de novo heterozygous 4-bp deletion (c.2853_2856delTCAG, NM_006306) in exon 18 of the SMC1A gene, resulting in a frameshift and premature termination (Ser951ArgfsTer12). The mutation was found by whole-exome sequencing and confirmed by Sanger sequencing. X-inactivation studies in peripheral blood cells showed a skewed pattern (93:7), although it was not clear if the mutant allele was expressed. Additional functional studies of the variant and studies of patient cells were not performed. The patient had onset of generalized tonic-clonic seizures at 4 months of age. The seizures became increasingly difficult to control, and she showed severe developmental regression and stagnation with inability to walk or speak.


.0009 DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 85 WITH MIDLINE BRAIN DEFECTS

SMC1A, 4-BP DUP, 3549GGCC
  
RCV000202430...

In a 3-year-old girl (patient B) with developmental and epileptic encephalopathy-85 and slight thinning of the corpus callosum (DEE85; 301044), Goldstein et al. (2015) identified a de novo heterozygous 4-bp duplication (c.3549_3552dupGGCC, NM_006306) in exon 24 of the SMC1A gene, resulting in a frameshift and premature termination (Ile1185GlyfsTer23). The mutation was found by whole-exome sequencing and confirmed by Sanger sequencing. X-inactivation studies in peripheral blood cells showed a random pattern. Additional functional studies of the variant and studies of patient cells were not performed. The patient had onset of tonic-clonic movements at 17 months of age following earlier global developmental delay.


.0010 DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 85 WITHOUT MIDLINE BRAIN DEFECTS

SMC1A, 1-BP DEL, NT2364
  
RCV001072126

In a 46-year-old woman (patient 1) with developmental and epileptic encephalopathy-85 (DEE85; 301044) without midline brain defects, Jansen et al. (2016) identified a de novo heterozygous 1-bp deletion (c.2364del, NM_006306) in the SMC1A gene, predicted to result in a frameshift and premature termination (Asn788LysfsTer10). The mutation was found by whole-genome sequencing and was predicted to result in nonsense-mediated mRNA decay and a loss of function. X-inactivation studies showed a random pattern. Additional functional studies were not performed. The patient had severe global developmental delay and onset of intractable seizures at 9 months of age.


.0011 DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 85 WITHOUT MIDLINE BRAIN DEFECTS

SMC1A, GLU733TER
  
RCV001072127

In an 8-year-old girl (patient 4) with developmental and epileptic encephalopathy-85 (DEE85; 301044) without midline brain defects, Symonds et al. (2017) identified a de novo heterozygous c.2197G-T transversion (c.2197G-T, NM_006306) in the SMC1A gene, predicted to result in a glu733-to-ter (E733X) substitution. The mutation was found by exome sequencing. Functional studies of the variant and studies of patient cells were not performed, although X-inactivation studies showed a normal pattern. The patient had onset of focal and generalized seizures that were difficult to control at 5 months of age.


.0012 DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 85 WITH OR WITHOUT MIDLINE BRAIN DEFECTS

SMC1A, 1-BP DEL, 2477A
  
RCV001072128

In 2 sisters (patients 8 and 9) with developmental and epileptic encephalopathy-85 (DEE85; 301044), Symonds et al. (2017) identified a de novo heterozygous 1-bp deletion (c.2477delA, NM_006306) in the SMC1A gene, predicted to result in a frameshift and premature termination. Functional studies of the variant and studies of patient cells were not performed. One sister had normal brain imaging and was less severely affected; she had onset of seizures at about 28 months of age. The other sister showed semilobar holoprosencephaly on brain imaging. She had onset of seizures in the first month of life and died at age 11 months. These findings demonstrated phenotypic variability even within the same family.


.0013 DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 85 WITH MIDLINE BRAIN DEFECTS

SMC1A, GLN1039TER
  
RCV001072129

In a girl (patient 10) who died at age 9 years with developmental and epileptic encephalopathy-85 with abnormal corpus callosum (DEE85; 301044), Symonds et al. (2017) identified a de novo heterozygous c.3115C-T transition (c.3115C-T, NM_006306) in the SMC1A gene, predicted to result in a gln1039-to-ter (Q1039X) substitution. Functional studies of the variant and studies in patient cells were not performed, but X-inactivation studies showed a skewed ratio (76:24). The patient had onset of intractable tonic-clonic seizures at 2 months of age.


.0014 DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 85 WITH MIDLINE BRAIN DEFECTS

SMC1A, ARG895GLY
  
RCV001072130

In a 6-year-old girl (patient 9) with developmental and epileptic encephalopathy-85 with semilobar holoprosencephaly (DEE85; 301044), Kruszka et al. (2019) identified a de novo heterozygous c.2683C-G transversion (chrX.53,423,417G-C, GRCh37) in the SMC1A gene, resulting in an arg895-to-gly (R895G) substitution at a conserved residue in the second coiled-coil domain. Functional studies of the variant and studies of patient cells were not performed, but the authors postulated either a loss-of-function or a dominant-negative effect. The patient had early-onset seizures.


.0015 DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 85 WITH MIDLINE BRAIN DEFECTS

SMC1A, 1-BP DEL, 2394A
  
RCV001072131

In a 3-year-old girl (patient 10) with developmental and epileptic encephalopathy-85 with semilobar holoprosencephaly (DEE85; 301044), Kruszka et al. (2019) identified a de novo heterozygous 1-bp deletion (c.2394delA; chrX.53,430,524delA) in the SMC1A gene, predicted to result in a frameshift and premature termination (Lys798AsnfsTer3). Functional studies of the variant and studies of patient cells were not performed. The patient had early-onset seizures.


REFERENCES

  1. Brown, C. J., Miller, A. P., Carrel, L., Rupert, J. L., Davies, K. E., Willard, H. F. The DXS423E gene in Xp11.21 escapes X chromosome inactivation. Hum. Molec. Genet. 4: 251-255, 1995. [PubMed: 7757075, related citations] [Full Text]

  2. Davidson, I. F., Bauer, B., Goetz, D., Tang, W., Wutz, G., Peters, J. M. DNA loop extrusion by human cohesin. Science 366: 1338-1345, 2019. [PubMed: 31753851, related citations] [Full Text]

  3. Deardorff, M. A., Kaur, M., Yaeger, D., Rampuria, A., Korolev, S., Pie, J., Gil-Rodriguez, C., Arnedo, M., Loeys, B., Kline, A. D., Wilson, M., Lillquist, K., Siu, V., Ramos, F. J., Musio, A., Jackson, L. S., Dorsett, D., Krantz, I. D. Mutations in cohesin complex members SMC3 and SMC1A cause a mild variant of Cornelia de Lange syndrome with predominant mental retardation. Am. J. Hum. Genet. 80: 485-494, 2007. [PubMed: 17273969, images, related citations] [Full Text]

  4. Egemen, A., Ulger, Z., Ozkinay, F., Gulen, F., Cogulu, O. A de novo t(X;8)(p11.2;q24.3) demonstrating Cornelia de Lange syndrome phenotype. Genet. Counsel. 16: 27-30, 2005. [PubMed: 15844775, related citations]

  5. Goldstein, J. H. R., Tim-aroon, T., Shieh, J., Merrill, M., Deeb, K. K., Zhang, S., Bass, N. E., Bedoyan, J. K. Novel SMC1A frameshift mutations in children with developmental delay and epilepsy. Europ. J. Med. Genet. 58: 562-568, 2015. [PubMed: 26386245, related citations] [Full Text]

  6. Gruber, S., Haering, C. H., Nasmyth, K. Chromosomal cohesin forms a ring. Cell 112: 765-777, 2003. [PubMed: 12654244, related citations] [Full Text]

  7. Haering, C. H., Farcas, A.-M., Arumugam, P., Metson, J., Nasmyth, K. The cohesin ring concatenates sister DNA molecules. Nature 454: 297-301, 2008. [PubMed: 18596691, related citations] [Full Text]

  8. Hoppman-Chaney, N., Jang, J. S., Jen, J., Babovic-Vuksanovic, D., Hodge, J. C. In-frame multi-exon deletion of SMC1A in a severely affected female with Cornelia de Lange syndrome. Am. J. Med. Genet. 158A: 193-198, 2012. [PubMed: 22106055, related citations] [Full Text]

  9. Jansen, S., Kleefstra, T., Willemsen, M. H., de Vries, P., Pfundt, R., Hehir-Kwa, J. Y., Gilissen, C., Veltman, J. A., de Vries, B. B. A., Vissers, L. E. L. M. De novo loss-of-function mutations in X-linked SMC1A cause severe ID and therapy-resistant epilepsy in females: expanding the phenotypic spectrum. Clin. Genet. 90: 413-419, 2016. [PubMed: 26752331, related citations] [Full Text]

  10. Kagey, M. H., Newman, J. J., Bilodeau, S., Zhan, Y., Orlando, D. A., van Berkum, N. L., Ebmeier, C. C., Goossens, J., Rahl, P. B., Levine, S. S., Taatjes, D. J., Dekker, J., Young, R. A. Mediator and cohesin connect gene expression and chromatin architecture. Nature 467: 430-435, 2010. Note: Erratum: Nature 472: 247 only, 2011. [PubMed: 20720539, images, related citations] [Full Text]

  11. Kruszka, P., Berger, S. I., Casa, V., Dekker, M. R., Gaesser, J., Weiss, K., Martinez, A. F., Murdock, D. R, Louie, R. J., Prijoles, E. J., Lichty, A. W., Brouwer, O. F., and 23 others. Cohesin complex-associated holoprosencephaly. Brain 142: 2631-2643, 2019. [PubMed: 31334757, related citations] [Full Text]

  12. Lebrun, N., Lebon, S., Jeannet, P.-Y., Jacquemont, S., Billuart, P., Bienvenu, T. Early-onset encephalopathy with epilepsy associated with a novel splice site mutation in SMC1A. Am. J. Med. Genet. 167A: 3076-3081, 2015. [PubMed: 26358754, related citations] [Full Text]

  13. Limongelli, G., Russo, S., Digilio, M. C., Masciadri, M., Pacileo, G., Fratta, F., Martone, F., Maddaloni, V., D'Alessandro, R., Calabro, P., Russo, M. G., Calabro, R., Larizza, L. Hypertrophic cardiomyopathy in a girl with Cornelia de Lange syndrome due to mutation in SMC1A. (Letter) Am. J. Med. Genet. 152A: 2127-2129, 2010. [PubMed: 20635401, related citations] [Full Text]

  14. Musio, A., Montagna, C., Mariani, T., Tilenni, M., Focarelli, M. L., Brait, L., Indino, E., Benedetti, P. A., Chessa, L., Albertini, A., Ried, T., Vezzoni, P. SMC1 involvement in fragile site expression. Hum. Molec. Genet. 14: 525-533, 2005. [PubMed: 15640246, related citations] [Full Text]

  15. Musio, A., Selicorni, A., Focarelli, M. L., Gervasini, C., Milani, D., Russo, S., Vezzoni, P., Larizza, L. X-linked Cornelia de Lange syndrome owing to SMC1L1 mutations. Nature Genet. 38: 528-530, 2006. [PubMed: 16604071, related citations] [Full Text]

  16. Patel, C. A., Ghiselli, G. Hinderin, a five-domains protein including coiled-coil motifs that binds to SMC3. BMC Cell Biol. 6: 3, 2005. Note: Electronic Article. [PubMed: 15656913, images, related citations] [Full Text]

  17. Revenkova, E., Focarelli, M. L., Susani, L., Paulis, M., Bassi, M. T., Mannini, L., Frattini, A., Delia, D., Krantz, I., Vezzoni, P., Jessberger, R., Musio, A. Cornelia de Lange syndrome mutations in SMC1A or SMC3 affect binding to DNA. Hum. Molec. Genet. 18: 418-427, 2009. [PubMed: 18996922, images, related citations] [Full Text]

  18. Rocques, P. J., Clark, J., Ball, S., Crew, J., Gill, S., Christodoulou, Z., Borts, R. H., Louis, E. J., Davies, K. E., Cooper, C. S. The human SB1.8 gene (DXS423E) encodes a putative chromosome segregation protein conserved in lower eukaryotes and prokaryotes. Hum. Molec. Genet. 4: 243-249, 1995. [PubMed: 7757074, related citations] [Full Text]

  19. Sultana, R., Adler, D. A., Edelhoff, S., Carrel, L., Lee, K., Chapman, V. C., Willard, H. F., Disteche, C. M. The mouse Sb1.8 gene located at the distal end of the X chromosome is subject to X inactivation. Hum. Molec. Genet. 4: 257-263, 1995. [PubMed: 7757076, related citations] [Full Text]

  20. Sumara, I., Vorlaufer, E., Gieffers, C., Peters, B. H., Peters, J.-M. Characterization of vertebrate cohesin complexes and their regulation in prophase. J. Cell Biol. 151: 749-761, 2000. [PubMed: 11076961, images, related citations] [Full Text]

  21. Symonds, J. D., Joss, S., Metcalfe, K. A., Somarathi, S., Cruden, J., Devlin, A. M., Donaldson, A., DiDonato, N., Fitzpatrick, D., Kaiser, F. J., Lampe, A. K., Lees, M. M., and 13 others. Heterozygous truncation mutations of the SMC1A gene cause a severe early onset epilepsy with cluster seizures in females: detailed phenotyping of 10 new cases. Epilepsia 58: 565-575, 2017. [PubMed: 28166369, related citations] [Full Text]


Ada Hamosh - updated : 05/06/2020
Cassandra L. Kniffin - updated : 04/10/2020
Patricia A. Hartz - updated : 07/24/2015
Cassandra L. Kniffin - updated : 2/16/2012
Cassandra L. Kniffin - updated : 1/11/2011
Ada Hamosh - updated : 10/7/2010
George E. Tiller - updated : 7/31/2009
Ada Hamosh - updated : 8/12/2008
George E. Tiller - updated : 1/3/2008
Victor A. McKusick - updated : 2/8/2007
Victor A. McKusick - updated : 4/26/2006
Stylianos E. Antonarakis - updated : 4/14/2003
Paul J. Converse - updated : 11/16/2001
Creation Date:
Victor A. McKusick : 3/14/1996
carol : 12/01/2020
ckniffin : 11/25/2020
alopez : 05/06/2020
carol : 04/16/2020
carol : 04/15/2020
carol : 04/14/2020
ckniffin : 04/10/2020
carol : 03/19/2019
mgross : 07/24/2015
carol : 2/21/2012
ckniffin : 2/16/2012
alopez : 6/10/2011
wwang : 2/1/2011
ckniffin : 1/11/2011
alopez : 10/8/2010
alopez : 10/8/2010
terry : 10/7/2010
wwang : 8/13/2009
terry : 7/31/2009
alopez : 8/25/2008
terry : 8/12/2008
alopez : 3/20/2008
terry : 3/7/2008
wwang : 1/14/2008
terry : 1/3/2008
carol : 12/5/2007
alopez : 2/13/2007
terry : 2/8/2007
carol : 8/8/2006
wwang : 5/4/2006
wwang : 4/27/2006
terry : 4/26/2006
alopez : 4/6/2005
mgross : 5/25/2004
mgross : 5/25/2004
ckniffin : 3/12/2004
mgross : 4/14/2003
mgross : 11/16/2001
alopez : 8/14/2000
alopez : 8/14/2000
mark : 3/14/1996
mark : 3/14/1996

* 300040

STRUCTURAL MAINTENANCE OF CHROMOSOMES 1A; SMC1A


Alternative titles; symbols

SMC1-ALPHA
STRUCTURAL MAINTENANCE OF CHROMOSOMES 1-LIKE 1; SMC1L1
SMC1
DXS423E
KIAA0178


HGNC Approved Gene Symbol: SMC1A

SNOMEDCT: 55016009;  


Cytogenetic location: Xp11.22     Genomic coordinates (GRCh38): X:53,374,149-53,422,728 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
Xp11.22 Cornelia de Lange syndrome 2 300590 X-linked dominant 3
Developmental and epileptic encephalopathy 85, with or without midline brain defects 301044 X-linked dominant 3

TEXT

Description

Eukaryotic sister chromatids remain connected from the time of synthesis until they are separated in anaphase. This cohesion depends on a complex of proteins known as cohesins. In vertebrates, unlike in yeast, the cohesins dissociate from chromosome arms earlier in M phase, during prophase. Small amounts of cohesin remain near the centromere until metaphase, with complete removal at the beginning of anaphase. Cohesin complexes contain SMC1, SCC1 (RAD21), SMC3 (606062), and either SA1 (STAG1; 604358) or SA2 (STAG2; 300826). The complexes, in turn, interact with PDS5 (see 613200), a protein implicated in chromosome cohesion, condensation, and recombination in yeast (summary by Sumara et al., 2000).


Cloning and Expression

Genetic marker SB1.8 (DXS423E) was originally identified as a cross-reacting clone during the screening of a human lymphocyte cDNA library with an oligonucleotide probe corresponding to the CYBB (300481) gene, which maps to Xp21.1. Rocques et al. (1995) found that the DXS423E gene encodes a protein of 1,233 amino acids that is 30% identical to the essential yeast protein SMC1 (structural maintenance of chromosomes-1), which is required for the segregation of chromosomes at mitosis. Both the human protein, called SB1.8, and SMC1 contain an N-terminal NTP-binding site, a central coiled-coil region, and a C-terminal helix-loop-helix domain, and both have structural features in common with the force-generating proteins myosin and kinesin. SB1.8 also exhibits regions of homology and overall structural similarity to protein 115p of the prokaryote Mycoplasma hyorhinis.

In mouse embryos, Kruszka et al. (2019) found expression of the Smc1a gene in anterior neural folds, the neuroectoderm, and adjacent mesenchyme of the developing brain. The findings suggested a role in forebrain patterning.


Gene Function

In yeast, the cohesin complex is essential for sister chromatid cohesion during mitosis. The Smc1 and Smc3 subunits are rod-shaped molecules with globular ABC-like ATPases at one end and dimerization domains at the other, connected by long coiled coils. Smc1 and Smc3 associate to form V-shaped heterodimers. Their ATPase heads are thought to be bridged by a third subunit, Scc1, creating a huge triangular ring that can trap sister DNA molecules. Gruber et al. (2003) studied whether cohesin forms such rings in vivo. Proteolytic cleavage of Scc1 by separase at the onset of anaphase triggers its dissociation from chromosomes. The authors showed that N- and C-terminal Scc1 cleavage fragments remain connected due to their association with different heads of a single Smc1/Smc3 heterodimer. Cleavage of the Smc3 coiled coil was sufficient to trigger cohesin release from chromosomes and loss of sister cohesion, consistent with a topologic association with chromatin.

By yeast 2-hybrid screening of a human fetal brain expression cDNA library using the hinge domain of SMC1 as bait, followed by immunoprecipitation analysis, Patel and Ghiselli (2005) found that SMC1 interacted with hinderin (KIAA1328; 616480). Hinderin did not interact with SMC3. Interaction of hinderin with SMC1 precluded dimerization of SMC1 and SMC3.

Musio et al. (2005) demonstrated that RNA interference (RNAi) of SMC1 was sufficient to induce fragile site expression in normal human fibroblasts. They showed that aphidicolin treatment led to an increase in SMC1 synthesis, SMC1 phosphorylation via an ATR (601215)-dependent pathway, and enhanced double-stranded break induction as visualized by immunohistochemical studies with phosphorylated H2AX (601772). Discrete nuclear foci were absent or very rare after 1 or 2 hours exposure to aphidicolin and/or RNAi of SMC1 but became more numerous and distinct after 6 hours. Musio et al. (2005) proposed that fragile sites might be viewed as an in vitro phenomenon originating from double-strand breaks formed because of stalled DNA replication that lasted too long to be rescued via the ATR/SMC1 axis, whereas in vivo, following an extreme replication block, rare cells could escape checkpoint mechanisms and enter mitosis with a defect in genome assembly, eventually leading to neoplastic transformation.

Cohesin's Scc1, Smc1, and Smc3 subunits form a tripartite ring structure, and it had been proposed that cohesin holds sister DNA molecules together by trapping them inside its ring. To test this, Haering et al. (2008) used site-specific crosslinking to create chemical connections at the 3 interfaces between the 3 constituent polypeptides of the ring, thereby creating covalently closed cohesin rings. As predicted by the ring entrapment model, this procedure produced dimeric DNA-cohesin structures that are resistant to protein denaturation. Haering et al. (2008) concluded that cohesin rings concatenate individual sister minichromosome DNA molecules.

Kagey et al. (2010) reported that Mediator (see MED8, 607956) and cohesin physically and functionally connect the enhancers and core promoters of active genes in murine embryonic stem cells. Mediator, a transcriptional coactivator, forms a complex with cohesin, which can form rings that connect 2 DNA segments. The cohesin-loading factor NIPBL (608667) is associated with Mediator-cohesin complexes, providing a means to load cohesin at promoters. DNA looping is observed between the enhancers and promoters occupied by Mediator and cohesin. Mediator and cohesin co-occupy different promoters in different cells, thus generating cell type-specific DNA loops linked to the gene expression program of each cell.

Using biochemical reconstitution, Davidson et al. (2019) found that single human cohesin complexes form DNA loops symmetrically at rates up to 2.1 kilobase pairs per second. Loop formation and maintenance depend on cohesin's ATPase activity and on NIPBL-MAU2 (614560), but not on topologic entrapment of DNA by cohesin (components include SMC3, 606062; SMC1A; STAG1, 604358; and STAG2, 300826). During loop formation, cohesin and NIPBL-MAU2 reside at the base of loops, which indicates that they generate loops by extrusion. Davidson et al. (2019) concluded that their results showed that cohesin and NIPBL-MAU2 form an active holoenzyme that interacts with DNA either pseudotopologically or nontopologically to extrude genomic interphase DNA into loops.


Mapping

Rocques et al. (1995) showed that the DXS423E gene maps to a cosmid contig that lies centromeric to the OATL2 locus (see 258870) at Xp11.2. Brown et al. (1995) showed that SMC1 escapes X-chromosome inactivation. SMC1 and XE169 (314690) were thought to define a new region in the proximal short arm of the X chromosome that escapes X inactivation. The corresponding gene in the mouse, Sb1.8, is located at the distal end of the X chromosome and is subject to X inactivation (Sultana et al., 1995).


Molecular Genetics

Cornelia de Lange Syndrome 2

A female with a Cornelia de Lange syndrome (122470) phenotype carrying an apparently balanced X;8 translocation involving the Xp11.2 band, to which the SMC1L1 gene maps, was described by Egemen et al. (2005). The findings were consistent with the location of the SMC1L1 gene at Xp11.2 and the involvement of that gene in X-linked Cornelia de Lange syndrome-2 (CDLS2; 300590).

Musio et al. (2006) recruited 53 unrelated and 4 related individuals with a diagnosis of Cornelia de Lange syndrome, encompassing the entire spectrum of phenotypes. They found pathogenic NIPBL (608667) mutations in 24 of them, whereas the remaining 33 cases did not bear any NIPBL mutation. Of these 33 individuals, there was only 1 instance of familial occurrence, with 2 male sibs, their mother, and a first cousin affected. Involvement of the NIPBL gene was excluded in this family, but the affected individuals were found to carry a 3-bp deletion in the SMC1L1 gene (300040.0001). In addition, a sporadic case was found to have a de novo missense mutation in the SMC1L1 gene (300040.0002).

Deardorff et al. (2007) identified 14 additional SMC1A mutations in patients with a mild variant of Cornelia de Lange syndrome with predominant mental retardation. Analysis of the mutant SMC1A proteins indicated that they were likely to produce functional cohesin complexes; however, Deardorff et al. (2007) posited that they mutations may alter their chromosome binding dynamics. Ten of 14 SMC1A-mutation-positive individuals with CDLS identified by Deardorff et al. (2007) were female. Furthermore, their series included similarly affected male and female probands, implying an X-linked dominant mode of expression. Several males were rather mildly affected and no more severely affected than many of the SMC1A mutation-positive females. Since the SMC1A gene escapes X inactivation (Brown et al., 1995), it is likely that the mechanism in affected females is due to a dominant-negative effect of the altered protein and less likely that it is due to decreased protein levels or skewed X inactivation. Consistent with this dominant-negative effect on cohesin, Deardorff et al. (2007) described a single amino acid deletion mutation in the SMC3 gene underlying a variant Cornelia de Lange syndrome (606062.0001). The data indicated that SMC3 and SMC1A mutations contribute to approximately 5% of cases of Cornelia de Lange syndrome, result in a consistently mild phenotype with absence of major structural anomalies typically associated with CDLS, such as those of the limbs, and, in some instances, result in a phenotype that approaches that of apparently nonsyndromic mental retardation. Deardorff et al. (2007) suggested that it may be found that additional 'cohesinopathies' result from perturbation of the more than 15 additional components of this complex that had yet to be associated with human disorders.

Developmental and Epileptic Encephalopathy 85 with or without Midline Brain Defects

In a 7-year-old girl, born of unrelated Portuguese parents, with developmental and epileptic encephalopathy-85 with midline brain defects manifest as thin corpus callosum (DEE85; 301044), Lebrun et al. (2015) identified a de novo heterozygous splice site mutation in the SMC1A gene (300040.0007). The mutation was found by exome sequencing and confirmed by Sanger sequencing. Analysis of patient fibroblasts showed the presence of only the mutant transcript, which was significantly reduced compared to controls, suggesting nonsense-mediated mRNA decay and a loss of function.

In 2 unrelated girls with DEE85, Goldstein et al. (2015) identified de novo heterozygous frameshift mutations in the SMC1A gene (300040.0008 and 300040.0009). The mutations were found by whole-exome sequencing and confirmed by Sanger sequencing. X-inactivation studies in peripheral blood cells showed a skewed pattern (93:7) in 1 patient, but a random pattern in the other. Additional functional studies of the variants and studies of patient cells were not performed.

In 2 unrelated females with DEE85, Jansen et al. (2016) identified de novo heterozygous loss of function (LOF) mutations in the SMC1A gene (see, e.g., 300040.0010). The mutations were found by whole-genome sequencing. X-inactivation studies in 1 patient showed a random pattern, whereas it was skewed in the other patient (85:15). Additional functional studies were not performed. Jansen et al. (2016) concluded that the de novo LOF mutations in the SMC1A gene in females cause an abnormal dosage effect and that the resulting phenotype is distinct from CDLS2. The authors further suggested that LOF mutations in males may be embryonic lethal.

In 10 unrelated females with DEE85, Symonds et al. (2017) identified de novo heterozygous nonsense, frameshift, or splice site mutations in the SMC1A gene (see, e.g., 300040.0011-300040.0013). Functional studies of the variants were not performed. X-inactivation studies, performed in some patients, had variable results: some showed a random pattern, whereas others had a skewed pattern. Symonds et al. (2017) speculated that if SMC1A escapes X inactivation, then haploinsufficiency is unlikely to be the causative mechanism; rather, the authors postulated a dominant-negative effect. The findings also suggested that complete SMC1A deficiency is embryonic lethal, as no males with such mutations have been reported.

In 5 unrelated girls (patients 7-11) with DEE85 and variable midline brain defects, including holoprosencephaly (HPE), Kruszka et al. (2019) identified de novo heterozygous mutations in the SMC1A gene (see, e.g., 300040.0014 and 300040.0015). All mutations except 1 were nonsense, frameshift, or splice site mutations, suggesting a loss-of-function effect; there was 1 missense mutation. Functional studies of the variants and studies of patient cells were not performed. Knockdown of the SMC1A gene in human neural progenitor cells resulted in upregulation of GLI2 (165230), ZIC2 (603073), and SMAD3 (603109) gene expression. Although the significance of these findings was unclear, it demonstrated that loss of SMC1A perturbs the expression of genes involved in HPE. The patients were part of a cohort of 277 individuals with HPE as well as gathered through collaborative efforts such as GeneMatcher.


ALLELIC VARIANTS 15 Selected Examples):

.0001   CORNELIA DE LANGE SYNDROME 2

SMC1A, 3-BP DEL, 2493CCA
SNP: rs1602407457, ClinVar: RCV000012438

Musio et al. (2006) identified a 3-bp deletion in the SMC1L1 gene in hemizygous state in 2 brothers and a maternal first cousin from a family with X-linked Cornelia de Lange syndrome (300590). The mother of the 2 brothers was heterozygous for the mutation and mildly affected. The deletion involved the third nucleotide of codon 831 and the first 2 nucleotides of codon 832, leading to deletion of gln832 and an asp831-to-glu (D831E) substitution. The mutation was not identified in over 600 control chromosomes.


.0002   CORNELIA DE LANGE SYNDROME 2

SMC1A, GLU493ALA
SNP: rs122454122, ClinVar: RCV000012439

In a male patient with sporadic X-linked Cornelia de Lange syndrome (300590), Musio et al. (2006) identified hemizygosity for a 1478A-C transversion in the SMC1L1 gene, resulting in an glu493-to-ala (E493A) substitution. The mutation was not identified in over 400 control chromosomes.

Revenkova et al. (2009) showed that E493A-mutant SMC1A affected the affinity of SMC hinge dimers for DNA. Mutated hinge dimers bound DNA with higher affinity than wildtype proteins. SMC1A-mutated Cornelia de Lange syndrome cell lines displayed genomic instability and sensitivity to ionizing radiation and interstrand crosslinking agents.


.0003   CORNELIA DE LANGE SYNDROME 2

SMC1A, 15-BP DEL, NT173
ClinVar: RCV000012440

In a boy with a milder variant of Cornelia de Lange syndrome (CDLS2; 300590), Deardorff et al. (2007) identified a 15-bp deletion in the SMC1A gene, resulting in deletion of 5 amino acids (V58_R62del). Features of CDLS included arched eyebrows and synophrys, anteverted nostrils, long and featureless philtrum, thin lips, downturned corners of the mouth, hearing loss, cutis marmorata, small hands and feet, proximally set thumbs, clinodactyly of fifth finger, and hirsutism. The child had psychomotor delay but was in mainstream second grade schooling. He had mild pulmonic stenosis and gastroesophageal reflux.


.0004   CORNELIA DE LANGE SYNDROME 2

SMC1A, ARG496HIS
SNP: rs122454123, ClinVar: RCV000012441, RCV001577833

In 2 sisters with a variant form of Cornelia de Lange syndrome (CDLS2; 300590), Deardorff et al. (2007) identified a 1487G-A transition in the SMC1A gene that resulted in an arg496-to-his substitution (R496H). Both had psychomotor delay and moderately severe mental retardation. One had pulmonic stenosis. Both had gastroesophageal reflux.

Revenkova et al. (2009) showed that R496H-mutant SMC1A affected the affinity of SMC hinge dimers for DNA. Mutated hinge dimers bound DNA with higher affinity than wildtype proteins. SMC1A-mutated Cornelia de Lange syndrome cell lines displayed genomic instability and sensitivity to ionizing radiation and interstrand crosslinking agents.


.0005   CORNELIA DE LANGE SYNDROME 2

SMC1A, ILE784THR
SNP: rs387906702, ClinVar: RCV000022820, RCV000441375, RCV000623393

In a 6-year-old girl with Cornelia de Lange syndrome (CDLS2; 300590), Limongelli et al. (2010) identified a de novo heterozygous 2351T-C transition in exon 15 of the SMC1A gene, resulting in an ile784-to-thr (I784T) substitution in a highly conserved residue in the coiled-coil domain. She had pre- and postnatal growth retardation, developmental delay, characteristic facial features, and concentric left ventricular hypertrophic cardiomyopathy.


.0006   CORNELIA DE LANGE SYNDROME 2

SMC1A, 8.152-KB DEL
ClinVar: RCV000022821

In a girl with a severe form of Cornelia de Lange syndrome-2 (CDLS2; 300590), Hoppman-Chaney et al. (2012) identified a de novo heterozygous 8.152-kb deletion encompassing exon 13 to intron 16 of the SMC1A gene, resulting in an in-frame deletion of 126 amino acids and insertion of 3 novel amino acids. The mutant mRNA was expressed, and Hoppman-Chaney et al. (2012) concluded that the mutant protein lacking the coiled-coil domain would act in a dominant-negative manner. The patient had dysmorphic facial features, microcephaly, poor growth, profound psychomotor retardation, holoprosencephaly, right hemihypertrophy, and mild distal limb anomalies. The patient was also mosaic for Turner syndrom, 45,X(7)/46,XX(23), which may have accounted for some additional features.


.0007   DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 85 WITH MIDLINE BRAIN DEFECTS

SMC1A, IVS11DS, G-T, +1
SNP: rs2075687835, ClinVar: RCV001072123

In a 7-year-old girl, born of unrelated Portuguese parents, with developmental and epileptic encephalopathy-85 and thin corpus callosum (DEE85; 301044), Lebrun et al. (2015) identified a de novo heterozygous G-to-T transversion (c.1911+1G-T) in intron 11 of the SMC1A gene, predicted to result in a splicing defect, frameshift, and premature termination (Thr638ValfsTer48). The mutation was found by exome sequencing and confirmed by Sanger sequencing. Analysis of patient fibroblasts showed the presence of only the mutant transcript, which was significantly reduced compared to controls, suggesting nonsense-mediated mRNA decay and a loss of function. The patient had onset of infantile spasms associated with hypsarrhythmia on EEG within the first month of life. She also had microcephaly, dysmorphic facial features, and severe developmental delay. Brain imaging showed a thin corpus callosum.


.0008   DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 85 WITH MIDLINE BRAIN DEFECTS

SMC1A, 4-BP DEL, 2853TCAG
SNP: rs863225458, ClinVar: RCV000202429, RCV000394437, RCV001072124

In a 4-year-old girl (patient A) with developmental and epileptic encephalopathy-85 (DEE85; 301044) with possible thickening of the insular cortex on brain imaging, Goldstein et al. (2015) identified a de novo heterozygous 4-bp deletion (c.2853_2856delTCAG, NM_006306) in exon 18 of the SMC1A gene, resulting in a frameshift and premature termination (Ser951ArgfsTer12). The mutation was found by whole-exome sequencing and confirmed by Sanger sequencing. X-inactivation studies in peripheral blood cells showed a skewed pattern (93:7), although it was not clear if the mutant allele was expressed. Additional functional studies of the variant and studies of patient cells were not performed. The patient had onset of generalized tonic-clonic seizures at 4 months of age. The seizures became increasingly difficult to control, and she showed severe developmental regression and stagnation with inability to walk or speak.


.0009   DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 85 WITH MIDLINE BRAIN DEFECTS

SMC1A, 4-BP DUP, 3549GGCC
SNP: rs863225459, ClinVar: RCV000202430, RCV001072125

In a 3-year-old girl (patient B) with developmental and epileptic encephalopathy-85 and slight thinning of the corpus callosum (DEE85; 301044), Goldstein et al. (2015) identified a de novo heterozygous 4-bp duplication (c.3549_3552dupGGCC, NM_006306) in exon 24 of the SMC1A gene, resulting in a frameshift and premature termination (Ile1185GlyfsTer23). The mutation was found by whole-exome sequencing and confirmed by Sanger sequencing. X-inactivation studies in peripheral blood cells showed a random pattern. Additional functional studies of the variant and studies of patient cells were not performed. The patient had onset of tonic-clonic movements at 17 months of age following earlier global developmental delay.


.0010   DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 85 WITHOUT MIDLINE BRAIN DEFECTS

SMC1A, 1-BP DEL, NT2364
SNP: rs2075680107, ClinVar: RCV001072126

In a 46-year-old woman (patient 1) with developmental and epileptic encephalopathy-85 (DEE85; 301044) without midline brain defects, Jansen et al. (2016) identified a de novo heterozygous 1-bp deletion (c.2364del, NM_006306) in the SMC1A gene, predicted to result in a frameshift and premature termination (Asn788LysfsTer10). The mutation was found by whole-genome sequencing and was predicted to result in nonsense-mediated mRNA decay and a loss of function. X-inactivation studies showed a random pattern. Additional functional studies were not performed. The patient had severe global developmental delay and onset of intractable seizures at 9 months of age.


.0011   DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 85 WITHOUT MIDLINE BRAIN DEFECTS

SMC1A, GLU733TER
SNP: rs2075681060, ClinVar: RCV001072127

In an 8-year-old girl (patient 4) with developmental and epileptic encephalopathy-85 (DEE85; 301044) without midline brain defects, Symonds et al. (2017) identified a de novo heterozygous c.2197G-T transversion (c.2197G-T, NM_006306) in the SMC1A gene, predicted to result in a glu733-to-ter (E733X) substitution. The mutation was found by exome sequencing. Functional studies of the variant and studies of patient cells were not performed, although X-inactivation studies showed a normal pattern. The patient had onset of focal and generalized seizures that were difficult to control at 5 months of age.


.0012   DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 85 WITH OR WITHOUT MIDLINE BRAIN DEFECTS

SMC1A, 1-BP DEL, 2477A
SNP: rs2075664229, ClinVar: RCV001072128

In 2 sisters (patients 8 and 9) with developmental and epileptic encephalopathy-85 (DEE85; 301044), Symonds et al. (2017) identified a de novo heterozygous 1-bp deletion (c.2477delA, NM_006306) in the SMC1A gene, predicted to result in a frameshift and premature termination. Functional studies of the variant and studies of patient cells were not performed. One sister had normal brain imaging and was less severely affected; she had onset of seizures at about 28 months of age. The other sister showed semilobar holoprosencephaly on brain imaging. She had onset of seizures in the first month of life and died at age 11 months. These findings demonstrated phenotypic variability even within the same family.


.0013   DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 85 WITH MIDLINE BRAIN DEFECTS

SMC1A, GLN1039TER
SNP: rs2075591576, ClinVar: RCV001072129

In a girl (patient 10) who died at age 9 years with developmental and epileptic encephalopathy-85 with abnormal corpus callosum (DEE85; 301044), Symonds et al. (2017) identified a de novo heterozygous c.3115C-T transition (c.3115C-T, NM_006306) in the SMC1A gene, predicted to result in a gln1039-to-ter (Q1039X) substitution. Functional studies of the variant and studies in patient cells were not performed, but X-inactivation studies showed a skewed ratio (76:24). The patient had onset of intractable tonic-clonic seizures at 2 months of age.


.0014   DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 85 WITH MIDLINE BRAIN DEFECTS

SMC1A, ARG895GLY
SNP: rs2075651835, ClinVar: RCV001072130

In a 6-year-old girl (patient 9) with developmental and epileptic encephalopathy-85 with semilobar holoprosencephaly (DEE85; 301044), Kruszka et al. (2019) identified a de novo heterozygous c.2683C-G transversion (chrX.53,423,417G-C, GRCh37) in the SMC1A gene, resulting in an arg895-to-gly (R895G) substitution at a conserved residue in the second coiled-coil domain. Functional studies of the variant and studies of patient cells were not performed, but the authors postulated either a loss-of-function or a dominant-negative effect. The patient had early-onset seizures.


.0015   DEVELOPMENTAL AND EPILEPTIC ENCEPHALOPATHY 85 WITH MIDLINE BRAIN DEFECTS

SMC1A, 1-BP DEL, 2394A
SNP: rs1569356555, ClinVar: RCV001072131

In a 3-year-old girl (patient 10) with developmental and epileptic encephalopathy-85 with semilobar holoprosencephaly (DEE85; 301044), Kruszka et al. (2019) identified a de novo heterozygous 1-bp deletion (c.2394delA; chrX.53,430,524delA) in the SMC1A gene, predicted to result in a frameshift and premature termination (Lys798AsnfsTer3). Functional studies of the variant and studies of patient cells were not performed. The patient had early-onset seizures.


REFERENCES

  1. Brown, C. J., Miller, A. P., Carrel, L., Rupert, J. L., Davies, K. E., Willard, H. F. The DXS423E gene in Xp11.21 escapes X chromosome inactivation. Hum. Molec. Genet. 4: 251-255, 1995. [PubMed: 7757075] [Full Text: https://doi.org/10.1093/hmg/4.2.251]

  2. Davidson, I. F., Bauer, B., Goetz, D., Tang, W., Wutz, G., Peters, J. M. DNA loop extrusion by human cohesin. Science 366: 1338-1345, 2019. [PubMed: 31753851] [Full Text: https://doi.org/10.1126/science.aaz3418]

  3. Deardorff, M. A., Kaur, M., Yaeger, D., Rampuria, A., Korolev, S., Pie, J., Gil-Rodriguez, C., Arnedo, M., Loeys, B., Kline, A. D., Wilson, M., Lillquist, K., Siu, V., Ramos, F. J., Musio, A., Jackson, L. S., Dorsett, D., Krantz, I. D. Mutations in cohesin complex members SMC3 and SMC1A cause a mild variant of Cornelia de Lange syndrome with predominant mental retardation. Am. J. Hum. Genet. 80: 485-494, 2007. [PubMed: 17273969] [Full Text: https://doi.org/10.1086/511888]

  4. Egemen, A., Ulger, Z., Ozkinay, F., Gulen, F., Cogulu, O. A de novo t(X;8)(p11.2;q24.3) demonstrating Cornelia de Lange syndrome phenotype. Genet. Counsel. 16: 27-30, 2005. [PubMed: 15844775]

  5. Goldstein, J. H. R., Tim-aroon, T., Shieh, J., Merrill, M., Deeb, K. K., Zhang, S., Bass, N. E., Bedoyan, J. K. Novel SMC1A frameshift mutations in children with developmental delay and epilepsy. Europ. J. Med. Genet. 58: 562-568, 2015. [PubMed: 26386245] [Full Text: https://doi.org/10.1016/j.ejmg.2015.09.007]

  6. Gruber, S., Haering, C. H., Nasmyth, K. Chromosomal cohesin forms a ring. Cell 112: 765-777, 2003. [PubMed: 12654244] [Full Text: https://doi.org/10.1016/s0092-8674(03)00162-4]

  7. Haering, C. H., Farcas, A.-M., Arumugam, P., Metson, J., Nasmyth, K. The cohesin ring concatenates sister DNA molecules. Nature 454: 297-301, 2008. [PubMed: 18596691] [Full Text: https://doi.org/10.1038/nature07098]

  8. Hoppman-Chaney, N., Jang, J. S., Jen, J., Babovic-Vuksanovic, D., Hodge, J. C. In-frame multi-exon deletion of SMC1A in a severely affected female with Cornelia de Lange syndrome. Am. J. Med. Genet. 158A: 193-198, 2012. [PubMed: 22106055] [Full Text: https://doi.org/10.1002/ajmg.a.34360]

  9. Jansen, S., Kleefstra, T., Willemsen, M. H., de Vries, P., Pfundt, R., Hehir-Kwa, J. Y., Gilissen, C., Veltman, J. A., de Vries, B. B. A., Vissers, L. E. L. M. De novo loss-of-function mutations in X-linked SMC1A cause severe ID and therapy-resistant epilepsy in females: expanding the phenotypic spectrum. Clin. Genet. 90: 413-419, 2016. [PubMed: 26752331] [Full Text: https://doi.org/10.1111/cge.12729]

  10. Kagey, M. H., Newman, J. J., Bilodeau, S., Zhan, Y., Orlando, D. A., van Berkum, N. L., Ebmeier, C. C., Goossens, J., Rahl, P. B., Levine, S. S., Taatjes, D. J., Dekker, J., Young, R. A. Mediator and cohesin connect gene expression and chromatin architecture. Nature 467: 430-435, 2010. Note: Erratum: Nature 472: 247 only, 2011. [PubMed: 20720539] [Full Text: https://doi.org/10.1038/nature09380]

  11. Kruszka, P., Berger, S. I., Casa, V., Dekker, M. R., Gaesser, J., Weiss, K., Martinez, A. F., Murdock, D. R, Louie, R. J., Prijoles, E. J., Lichty, A. W., Brouwer, O. F., and 23 others. Cohesin complex-associated holoprosencephaly. Brain 142: 2631-2643, 2019. [PubMed: 31334757] [Full Text: https://doi.org/10.1093/brain/awz210]

  12. Lebrun, N., Lebon, S., Jeannet, P.-Y., Jacquemont, S., Billuart, P., Bienvenu, T. Early-onset encephalopathy with epilepsy associated with a novel splice site mutation in SMC1A. Am. J. Med. Genet. 167A: 3076-3081, 2015. [PubMed: 26358754] [Full Text: https://doi.org/10.1002/ajmg.a.37364]

  13. Limongelli, G., Russo, S., Digilio, M. C., Masciadri, M., Pacileo, G., Fratta, F., Martone, F., Maddaloni, V., D'Alessandro, R., Calabro, P., Russo, M. G., Calabro, R., Larizza, L. Hypertrophic cardiomyopathy in a girl with Cornelia de Lange syndrome due to mutation in SMC1A. (Letter) Am. J. Med. Genet. 152A: 2127-2129, 2010. [PubMed: 20635401] [Full Text: https://doi.org/10.1002/ajmg.a.33486]

  14. Musio, A., Montagna, C., Mariani, T., Tilenni, M., Focarelli, M. L., Brait, L., Indino, E., Benedetti, P. A., Chessa, L., Albertini, A., Ried, T., Vezzoni, P. SMC1 involvement in fragile site expression. Hum. Molec. Genet. 14: 525-533, 2005. [PubMed: 15640246] [Full Text: https://doi.org/10.1093/hmg/ddi049]

  15. Musio, A., Selicorni, A., Focarelli, M. L., Gervasini, C., Milani, D., Russo, S., Vezzoni, P., Larizza, L. X-linked Cornelia de Lange syndrome owing to SMC1L1 mutations. Nature Genet. 38: 528-530, 2006. [PubMed: 16604071] [Full Text: https://doi.org/10.1038/ng1779]

  16. Patel, C. A., Ghiselli, G. Hinderin, a five-domains protein including coiled-coil motifs that binds to SMC3. BMC Cell Biol. 6: 3, 2005. Note: Electronic Article. [PubMed: 15656913] [Full Text: https://doi.org/10.1186/1471-2121-6-3]

  17. Revenkova, E., Focarelli, M. L., Susani, L., Paulis, M., Bassi, M. T., Mannini, L., Frattini, A., Delia, D., Krantz, I., Vezzoni, P., Jessberger, R., Musio, A. Cornelia de Lange syndrome mutations in SMC1A or SMC3 affect binding to DNA. Hum. Molec. Genet. 18: 418-427, 2009. [PubMed: 18996922] [Full Text: https://doi.org/10.1093/hmg/ddn369]

  18. Rocques, P. J., Clark, J., Ball, S., Crew, J., Gill, S., Christodoulou, Z., Borts, R. H., Louis, E. J., Davies, K. E., Cooper, C. S. The human SB1.8 gene (DXS423E) encodes a putative chromosome segregation protein conserved in lower eukaryotes and prokaryotes. Hum. Molec. Genet. 4: 243-249, 1995. [PubMed: 7757074] [Full Text: https://doi.org/10.1093/hmg/4.2.243]

  19. Sultana, R., Adler, D. A., Edelhoff, S., Carrel, L., Lee, K., Chapman, V. C., Willard, H. F., Disteche, C. M. The mouse Sb1.8 gene located at the distal end of the X chromosome is subject to X inactivation. Hum. Molec. Genet. 4: 257-263, 1995. [PubMed: 7757076] [Full Text: https://doi.org/10.1093/hmg/4.2.257]

  20. Sumara, I., Vorlaufer, E., Gieffers, C., Peters, B. H., Peters, J.-M. Characterization of vertebrate cohesin complexes and their regulation in prophase. J. Cell Biol. 151: 749-761, 2000. [PubMed: 11076961] [Full Text: https://doi.org/10.1083/jcb.151.4.749]

  21. Symonds, J. D., Joss, S., Metcalfe, K. A., Somarathi, S., Cruden, J., Devlin, A. M., Donaldson, A., DiDonato, N., Fitzpatrick, D., Kaiser, F. J., Lampe, A. K., Lees, M. M., and 13 others. Heterozygous truncation mutations of the SMC1A gene cause a severe early onset epilepsy with cluster seizures in females: detailed phenotyping of 10 new cases. Epilepsia 58: 565-575, 2017. [PubMed: 28166369] [Full Text: https://doi.org/10.1111/epi.13669]


Contributors:
Ada Hamosh - updated : 05/06/2020
Cassandra L. Kniffin - updated : 04/10/2020
Patricia A. Hartz - updated : 07/24/2015
Cassandra L. Kniffin - updated : 2/16/2012
Cassandra L. Kniffin - updated : 1/11/2011
Ada Hamosh - updated : 10/7/2010
George E. Tiller - updated : 7/31/2009
Ada Hamosh - updated : 8/12/2008
George E. Tiller - updated : 1/3/2008
Victor A. McKusick - updated : 2/8/2007
Victor A. McKusick - updated : 4/26/2006
Stylianos E. Antonarakis - updated : 4/14/2003
Paul J. Converse - updated : 11/16/2001

Creation Date:
Victor A. McKusick : 3/14/1996

Edit History:
carol : 12/01/2020
ckniffin : 11/25/2020
alopez : 05/06/2020
carol : 04/16/2020
carol : 04/15/2020
carol : 04/14/2020
ckniffin : 04/10/2020
carol : 03/19/2019
mgross : 07/24/2015
carol : 2/21/2012
ckniffin : 2/16/2012
alopez : 6/10/2011
wwang : 2/1/2011
ckniffin : 1/11/2011
alopez : 10/8/2010
alopez : 10/8/2010
terry : 10/7/2010
wwang : 8/13/2009
terry : 7/31/2009
alopez : 8/25/2008
terry : 8/12/2008
alopez : 3/20/2008
terry : 3/7/2008
wwang : 1/14/2008
terry : 1/3/2008
carol : 12/5/2007
alopez : 2/13/2007
terry : 2/8/2007
carol : 8/8/2006
wwang : 5/4/2006
wwang : 4/27/2006
terry : 4/26/2006
alopez : 4/6/2005
mgross : 5/25/2004
mgross : 5/25/2004
ckniffin : 3/12/2004
mgross : 4/14/2003
mgross : 11/16/2001
alopez : 8/14/2000
alopez : 8/14/2000
mark : 3/14/1996
mark : 3/14/1996