Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Mutations in SGOL1 cause a novel cohesinopathy affecting heart and gut rhythm

Abstract

The pacemaking activity of specialized tissues in the heart and gut results in lifelong rhythmic contractions. Here we describe a new syndrome characterized by Chronic Atrial and Intestinal Dysrhythmia, termed CAID syndrome, in 16 French Canadians and 1 Swede. We show that a single shared homozygous founder mutation in SGOL1, a component of the cohesin complex, causes CAID syndrome. Cultured dermal fibroblasts from affected individuals showed accelerated cell cycle progression, a higher rate of senescence and enhanced activation of TGF-β signaling. Karyotypes showed the typical railroad appearance of a centromeric cohesion defect. Tissues derived from affected individuals displayed pathological changes in both the enteric nervous system and smooth muscle. Morpholino-induced knockdown of sgol1 in zebrafish recapitulated the abnormalities seen in humans with CAID syndrome. Our findings identify CAID syndrome as a novel generalized dysrhythmia, suggesting a new role for SGOL1 and the cohesin complex in mediating the integrity of human cardiac and gut rhythm.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Synopsis of families and electrocardiographic features.
Figure 2: Cellular phenotypes of CAID.
Figure 3: Synopsis of the gastrointestinal histology of CAID.
Figure 4: Spatiotemporal expression and function of sgol1 in zebrafish.

Similar content being viewed by others

Accession codes

Accessions

NCBI Reference Sequence

References

  1. Rodriguez, R.D. & Schocken, D.D. Update on sick sinus syndrome, a cardiac disorder of aging. Geriatrics 45, 26–30, 33–36 (1990).

    CAS  PubMed  Google Scholar 

  2. Benson, D.W. et al. Congenital sick sinus syndrome caused by recessive mutations in the cardiac sodium channel gene (SCN5A). J. Clin. Invest. 112, 1019–1028 (2003).

    Article  CAS  Google Scholar 

  3. Laish-Farkash, A. et al. A novel mutation in the HCN4 gene causes symptomatic sinus bradycardia in Moroccan Jews. J. Cardiovasc. Electrophysiol. 21, 1365–1372 (2010).

    Article  Google Scholar 

  4. Milanesi, R., Baruscotti, M., Gnecchi-Ruscone, T. & DiFrancesco, D. Familial sinus bradycardia associated with a mutation in the cardiac pacemaker channel. N. Engl. J. Med. 354, 151–157 (2006).

    Article  CAS  Google Scholar 

  5. Schulze-Bahr, E. et al. Pacemaker channel dysfunction in a patient with sinus node disease. J. Clin. Invest. 111, 1537–1545 (2003).

    Article  CAS  Google Scholar 

  6. Holm, H. et al. A rare variant in MYH6 is associated with high risk of sick sinus syndrome. Nat. Genet. 43, 316–320 (2011).

    Article  CAS  Google Scholar 

  7. Mohler, P.J. et al. Ankyrin-B mutation causes type 4 long-QT cardiac arrhythmia and sudden cardiac death. Nature 421, 634–639 (2003).

    Article  CAS  Google Scholar 

  8. Mohler, P.J. et al. A cardiac arrhythmia syndrome caused by loss of ankyrin-B function. Proc. Natl. Acad. Sci. USA 101, 9137–9142 (2004).

    Article  CAS  Google Scholar 

  9. Gargiulo, A. et al. Filamin A is mutated in X-linked chronic idiopathic intestinal pseudo-obstruction with central nervous system involvement. Am. J. Hum. Genet. 80, 751–758 (2007).

    Article  CAS  Google Scholar 

  10. Deglincerti, A. et al. A novel locus for syndromic chronic idiopathic intestinal pseudo-obstruction maps to chromosome 8q23-q24. Eur. J. Hum. Genet. 15, 889–897 (2007).

    Article  CAS  Google Scholar 

  11. Siva, N. 1000 Genomes project. Nat. Biotechnol. 26, 256 (2008).

    Article  Google Scholar 

  12. Purcell, S. et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet. 81, 559–575 (2007).

    Article  CAS  Google Scholar 

  13. Seelow, D., Schuelke, M., Hildebrandt, F. & Nürnberg, P. HomozygosityMapper—an interactive approach to homozygosity mapping. Nucleic Acids Res. 37, W593–W599 (2009).

    Article  CAS  Google Scholar 

  14. Bouchard, G., Roy, R., Casgrain, B. & Hubert, M. Population files and database management: the BALSAC database and the INGRES/INGRID system. Hist. Mes. 4, 39–57 (1989).

    Article  CAS  Google Scholar 

  15. Thompson, E.A. Identity by descent: variation in meiosis, across genomes, and in populations. Genetics 194, 301–326 (2013).

    Article  CAS  Google Scholar 

  16. Gramley, F. et al. Atrial fibrosis and atrial fibrillation: the role of the TGF-β1 signaling pathway. Int. J. Cardiol. 143, 405–413 (2010).

    Article  Google Scholar 

  17. Fu, M., Lui, V.C.H., Sham, M.H., Pachnis, V. & Tam, P.K.H. Sonic hedgehog regulates the proliferation, differentiation, and migration of enteric neural crest cells in gut. J. Cell Biol. 166, 673–684 (2004).

    Article  CAS  Google Scholar 

  18. Sukegawa, A. et al. The concentric structure of the developing gut is regulated by Sonic hedgehog derived from endodermal epithelium. Development 127, 1971–1980 (2000).

    CAS  PubMed  Google Scholar 

  19. Tessadori, F. et al. Identification and functional characterization of cardiac pacemaker cells in zebrafish. PLoS ONE 7, e47644 (2012).

    Article  CAS  Google Scholar 

  20. Pater, E. et al. Distinct phases of cardiomyocyte differentiation regulate growth of the zebrafish heart. Development 136, 1633–1641 (2009).

    Article  Google Scholar 

  21. Kline, A.D. et al. Cornelia de Lange syndrome: clinical review, diagnostic and scoring systems, and anticipatory guidance. Am. J. Med. Genet. A. 143A, 1287–1296 (2007).

    Article  Google Scholar 

  22. Gillis, L.A. et al. NIPBL mutational analysis in 120 individuals with Cornelia de Lange syndrome and evaluation of genotype-phenotype correlations. Am. J. Hum. Genet. 75, 610–623 (2004).

    Article  CAS  Google Scholar 

  23. Deardorff, M.A. et al. RAD21 mutations cause a human cohesinopathy. Am. J. Hum. Genet. 90, 1014–1027 (2012).

    Article  CAS  Google Scholar 

  24. Deardorff, M.A. et al. HDAC8 mutations in Cornelia de Lange syndrome affect the cohesin acetylation cycle. Nature 489, 313–317 (2012).

    Article  CAS  Google Scholar 

  25. Musio, A. et al. X-linked Cornelia de Lange syndrome owing to SMC1L1 mutations. Nat. Genet. 38, 528–530 (2006).

    Article  CAS  Google Scholar 

  26. van der Lelij, P. et al. Warsaw breakage syndrome, a cohesinopathy associated with mutations in the XPD helicase family member DDX11/ChlR1. Am. J. Hum. Genet. 86, 262–266 (2010).

    Article  CAS  Google Scholar 

  27. Caburet, S. et al. Mutant cohesin in premature ovarian failure. N. Engl. J. Med. 370, 943–949 (2014).

    Article  CAS  Google Scholar 

  28. Yamagishi, Y., Sakuno, T., Shimura, M. & Watanabe, Y. Heterochromatin links to centromeric protection by recruiting shugoshin. Nature 455, 251–255 (2008).

    Article  CAS  Google Scholar 

  29. Heijman, J., Dewenter, M., El-Armouche, A. & Dobrev, D. Function and regulation of serine/threonine phosphatases in the healthy and diseased heart. J. Mol. Cell. Cardiol. 64, 90–98 (2013).

    Article  CAS  Google Scholar 

  30. Schmidt, D. et al. A CTCF-independent role for cohesin in tissue-specific transcription. Genome Res. 20, 578–588 (2010).

    Article  CAS  Google Scholar 

  31. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  Google Scholar 

  32. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  Google Scholar 

  33. Flicek, P. et al. Ensembl 2012. Nucleic Acids Res. 40, D84–D90 (2012).

    Article  CAS  Google Scholar 

  34. International HapMap Consortium. The International HapMap Project. Nature 426, 789–796 (2003).

  35. Gabriel, S.B. et al. The structure of haplotype blocks in the human genome. Science 296, 2225–2229 (2002).

    Article  CAS  Google Scholar 

  36. Rajeevan, H. et al. ALFRED: an allele frequency database for microevolutionary studies. Evol. Bioinform. Online 1, 1–10 (2005).

    Article  CAS  Google Scholar 

  37. Labuda, M. et al. Linkage disequilibrium analysis in young populations: pseudo-vitamin D–deficiency rickets and the founder effect in French Canadians. Am. J. Hum. Genet. 59, 633–643 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Labuda, D., Zietkiewicz, E. & Labuda, M. The genetic clock and the age of the founder effect in growing populations: a lesson from French Canadians and Ashkenazim. Am. J. Hum. Genet. 61, 768–771 (1997).

    Article  CAS  Google Scholar 

  39. Glessner, J.T., Li, J. & Hakonarson, H. ParseCNV integrative copy number variation association software with quality tracking. Nucleic Acids Res. 41, e64 (2013).

    Article  CAS  Google Scholar 

  40. Subirana, I., Diaz-Uriarte, R., Lucas, G. & Gonzalez, J.R. CNVassoc: association analysis of CNV data using R. BMC Med. Genomics 4, 47 (2011).

    Article  Google Scholar 

  41. MacCluer, J.W., VandeBerg, J.L., Read, B. & Ryder, O.A. Pedigree analysis by computer simulation. Zoo Biol. 5, 147–160 (1986).

    Article  Google Scholar 

  42. Itahana, K., Campisi, J. & Dimri, G.P. Methods to detect biomarkers of cellular senescence: the senescence-associated β-galactosidase assay. Methods Mol. Biol. 371, 21–31 (2007).

    Article  CAS  Google Scholar 

  43. Notarnicola, C. et al. The RNA-binding protein RBPMS2 regulates development of gastrointestinal smooth muscle. Gastroenterology 143, 687–697 (2012).

    Article  CAS  Google Scholar 

  44. Rouleau, C., Matécki, S., Kalfa, N., Costes, V. & de Santa Barbara, P. Activation of MAP kinase (ERK1/2) in human neonatal colonic enteric nervous system. Neurogastroenterol. Motil. 21, 207–214 (2009).

    Article  CAS  Google Scholar 

  45. Westerfield, M. The Zebrafish Book: A Guide for the Laboratory Use of Zebrafish Danio (Brachydanio) rerio (University of Oregon Press, Eugene, Oregon, USA, 2000).

  46. Thisse, C. & Thisse, B. High-resolution in situ hybridization to whole-mount zebrafish embryos. Nat. Protoc. 3, 59–69 (2008).

    Article  CAS  Google Scholar 

  47. Moorman, A.F., Houweling, A.C., de Boer, P.A. & Christoffels, V.M. Sensitive nonradioactive detection of mRNA in tissue sections: novel application of the whole-mount in situ hybridization protocol. J. Histochem. Cytochem. 49, 1–8 (2001).

    Article  CAS  Google Scholar 

  48. Chopra, S.S. et al. Voltage-gated sodium channels are required for heart development in zebrafish. Circ. Res. 106, 1342–1350 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are deeply indebted to all participating families and patients. We thank L. Gosselin, L. L'Écuyer, M.-F. Boudreault, C. Faure, F. LeDeist, L. D'Aoust and the McGill Genome Québec Innovation Centre (D. Vincent) for expert assistance. We gratefully acknowledge the contribution of all referring physicians and supporting staff to this project. We would like to thank J. Marcadier (clinical coordinator) and C. Beaulieu (project manager) for their contributions to the infrastructure of the FORGE Canada Consortium. The FORGE (Finding of Rare Disease Genes) Consortium Steering Committee includes Kym Boycott (University of Ottawa), Jan Friedman (University of British Columbia), Jacques Michaud (Université de Montréal), François Bernier (University of Calgary), Michael Brudno (University of Toronto), Bridget Fernandez (Memorial University), Bartha Knoppers (McGill University) and Steve Scherer (University of Toronto). FORGE Canada was funded by the Canadian government through Genome Canada, the Canadian Institutes of Health Research and the Ontario Genomics Institute (OGI-049). Additional funding was provided by Génome Québec, Genome British Columbia, Foundation Nussia and André Aisenstadt, La Fondation du Grand défi Pierre Lavoie, Fondation CHU Sainte-Justine, Fondation Leducq, Association des Pseudo Obstructions Intestinales Chroniques (France), the Netherlands Organization for Scientific Research (NWO) and ZonMw (91212086). G.A. holds a Senior Scholarship from the Fonds de Recherche du Québec–Santé and holds the Banque Nationale Research Chair in Cardiovascular Genetics. We thank G. Rouleau for sharing whole-exome data for control purposes and M. Samuels for critical reading of the manuscript.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

P.C., J.-M.C. and G.A. identified the novel CAID syndrome in the families. Functional experiments were performed by F.W., N.G., J.P., S.L., C.G., E.L., I.B., J.B., S.B., A.G., E.T., S.F., M.C., N.E.A., E.G.-M., S.H.Z., Y.S., M.J. and S.J.M.J. Further patient recruitment and enrollment was performed by P.C., J.-M.C., C.H., J.C., A.J., P.d.S.B., M.T., D.W.B. and G.A. C.P. performed and supervised the genotyping and the haplotype and population genetic analysis. G.R.H., G.A., J.P. and N.E.A. performed confocal microscopy for the cell lines and analyzed the data. S.B. and J.B. established the zebrafish model. C.G., J.P. and E.G.-M. performed the functional cell line experiments. Genealogical analysis was provided by M.J., D.L. and C.M. H.C.D. provided advice on the analysis of the functional data derived from the cell lines. P.C., C.P., J.B., S.J.M.J. and G.A. wrote the manuscript with contributions and input from all authors. G.A. designed and supervised the study.

Corresponding author

Correspondence to Gregor Andelfinger.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Membership of the Steering Committee for the Consortium is given in the Acknowledgments

Integrated supplementary information

Supplementary Figure 1 Representative manometries in two CAID patients.

(a) Antroduodenal manometry showing visceral neuropathy with abnormal response (normal amplitude, no phase III–like contractions) to IV erythromycin (antrum, channels 1–3; duodenum, channels 4 and 5; jejunum, channels 6 and 7). (b) Colonic manometry showing visceral myopathy with low-amplitude propagated contractions after stimulation (intracolonic bisacodyl) (right colon, channels 1 and 2; transverse, channels 3 and 4; sigmoid rectum, channels 5–7).

Supplementary Figure 2 Results of sequencing.

(a) Venn diagram indicating the number of genes with homozygous nonsynonymous variants in three sequenced patients. In parentheses are the numbers of genes after filtering for rare alleles (MAF < 1%). (b) Examples of Sanger sequencing results for the screening of the exon 1 c.67A>G mutation (red arrow) showing the wild-type allele (top), a heterozygous parent (middle) and a homozygous CAID patient (bottom). One-letter amino acid code is used for the translation of individual codons. (c) The lysine (K) at position 23 is highly conserved in all vertebrates.

Source data

Supplementary Figure 3 Results from homozygosity and haplotype mapping.

The results from homozygosity mapping1 in 12 French-Canadian CAID patients, 3 unaffected family members (family 4) and the Swedish patient are displayed. (a,b) Graphic depiction of homozygosity scores (a) across the genome and (b) for chromosome 3 providing mapping evidence for the SGOL1 locus in CAID. (c) Reconstruction of the founder haplotype in the SGOL1 region based on the largest window of consecutive homozygous variant calls obtained from high-density genotyping. The haplotype is displayed on the UCSC Genome Browser, showing that a 650-kb interval overlapping the SGOL1 locus is identical by descent. Graphic depiction of haplotypes on the UCSC Genome Browser for the region between 20 Mb and 20.65 Mb on chromosome 3 harboring the following four genes: SGOL1, RAB5A, C3orf48 and PCAF.

Supplementary Figure 4 Haplotype gradients.

(a) The distribution of haplotype frequencies across a world map highlights the absence of the disease-associated haplotype outside of Europe and North America. The haplotype gradients follow a migration pattern from northern European populations across the Atlantic. (b) For simplicity, a set of seven rare tagging SNPs for three haplotype combinations of different length is shown. Recombination breaks down the haplotypes over time within a population2. The presence of the longest haplotype in northern European populations and in North America combined with the absence of this haplotype in southern Europeans undermines the idea of a transatlantic founder effect. (c) Overview of allele frequencies and genotype counts for the rare tagging SNPs from the 1000 Genomes Project panel. Colors highlight the higher frequencies of variants in the European population compared to the Asian and African populations.

Source data

Supplementary Figure 5 Genealogical analysis.

(a) The genealogy of the French-Canadian CAID patients linking all of them to their most likely founder couple married in France in 1620. (b) The corresponding genealogy of only one of the patients, illustrating details of the familial relationships along generations. The proposed founder couple is more than 100 times more likely to be the founder than other potential founders (posterior probability P = 0.993, estimated by gene dropping). Duplicated individuals are indicated by dashed lines. (c) Haplotype alignment. A schematic representation of the haplotypes around the CAID mutation (vertical line) found in nine patients, each representing an independent nuclear family, compared with two haplotypes for the Swedish patient. The consensus haplotype is represented by a solid line that is interrupted by empty spaces representing recombination break points. Notably, the Swedish patient shares about 0.5 cM with the Quebec haplotypes. We estimated the age of the founder haplotype to be between 9 and 17 generations, given proportions of 78% and 50% for the non-recombined mutation-carrying haplotypes at lengths of 0.28 and 0.41 cM, respectively. This is in line with the genealogical evidence pointing to the founder couple arriving in Nouvelle France around 1620 (ref. 3).

Supplementary Figure 6 Confocal analysis of SGOL1 during mitosis.

Comparison of a control wild-type cell line and two cell lines homozygous for the SGOL1 K23E mutant is shown. SGOL1 K23E localizes correctly to the centromeric regions during pro-metaphase and anaphase but retains an abnormal cytosolic localization pattern. Left: green, SGOL1 staining. Right: green and blue, SGOL1 plus Hoechst. Scale bar, 10 µm.

Supplementary Figure 7 Quantitative analysis of confocal microscopy.

Quantitative analysis of the distribution of nuclear SGOL1 pixel intensities reveals altered SGOL1 organization in fibroblasts from homozygous CAID patients (red) compared to controls (blue). Data are the mean ± s.e.m. from 12 cells from 3 individuals per genotype. In control nuclei, the distribution partially overlaps that of adjacent cell-free regions (background, green), reflecting nuclear domains of negligible SGOL1 signal intensity among domains of higher intensity. In patient nuclei, the SGOL1 (K23E) intensity distribution does not overlap the background, showing that, in fibroblasts from three different patients, nuclear SGOL1 is reproducibly delocalized compared to three different controls, consistent with the more diffuse staining patterns observed.

Supplementary Figure 8 SGOL1 expression in normal human colon.

SGOL1 expression in normal human colon. Immunohistochemistry shows that SGOL1 expression in the intestinal wall is very strong in enteric neuronal cells and interstitial cells of Cajal networks (black and red arrows, respectively). SGOL1 is expressed to a lower extent in smooth muscle cells. Antibodies used (left to right): SGOL1, TMEM16A (a calcium channel expressed in the network of interstitial cells of Cajal), TUJ1 (neuron-specific β III tubulin, a neuronal marker expressed in intestinal ganglial cells) and αSMA (α smooth muscle actin, a smooth muscle cell marker).

Supplementary Figure 9 Antisense morpholino knockdown of sgol1 analyzed at 3 d.p.f.

(a) Schematic of the morpholino target site location in the sgol1 gene transcript. (b) Representative images of control and sgol1 morpholino-injected embryos (3 d.p.f.). Arrows point to cardiac edema. (c) Analysis of cardiac cycle length by high-speed video imaging (n = 15 per group). *P < 0.05, **P < 0.01.

Source data

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9 and Supplementary Note. (PDF 6175 kb)

Supplementary Table 1

Summary of patient characteristics. (XLSX 10 kb)

Supplementary Table 2

Full characterization of patient cohort. (XLSX 15 kb)

Supplementary Table 3

In silico prediction of the deleterious SGOL1 mutation. (XLSX 12 kb)

Supplementary Table 4

Primer design and PCR. (XLSX 10 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chetaille, P., Preuss, C., Burkhard, S. et al. Mutations in SGOL1 cause a novel cohesinopathy affecting heart and gut rhythm. Nat Genet 46, 1245–1249 (2014). https://doi.org/10.1038/ng.3113

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3113

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing