Skip to main content
Log in

X-linked genes exhibit miR6891-5p-regulated skewing in Sjögren’s syndrome

  • Original Article
  • Published:
Journal of Molecular Medicine Aims and scope Submit manuscript

Abstract

Many autoimmune diseases exhibit a strikingly increased prevalence in females, with primary Sjögren’s syndrome (pSS) being the most female-predominant example. However, the molecular basis underlying the female-bias in pSS remains elusive. To address this knowledge gap, we performed genome-wide, allele-specific profiling of minor salivary gland-derived mesenchymal stromal cells (MSCs) from pSS patients and control subjects, and detected major differences in the regulation of X-linked genes. In control female MSCs, X-linked genes were expressed from both paternal and maternal X chromosomes with a median paternal ratio of ~ 0.5. However, in pSS female MSCs, X-linked genes exhibited preferential expression from one of the two X chromosomes. Concomitantly, pSS MSCs showed decrease in XIST levels and reorganization of H3K27me3+ foci in the nucleus. Moreover, the HLA-locus-expressed miRNA miR6891-5p was decreased in pSS MSCs. miR6891-5p inhibition in control MSCs caused XIST dysregulation, ectopic silencing, and allelic skewing. Allelic skewing was accompanied by the mislocation of protein products encoded by the skewed genes, which was recapitulated by XIST and miR6891-5p disruption in control MSCs. Our data reveal X skewing as a molecular hallmark of pSS and highlight the importance of restoring X-chromosomal allelic balance for pSS treatment.

Key messages

  • X-linked genes exhibit skewing in primary Sjögren’s syndrome (pSS).

  • X skewing in pSS associates with alterations in H3K27me3 deposition.

  • pSS MSCs show decreased levels of miR6891-5p, a HLA-expressed miRNA.

  • miR6891-5p inhibition causes H3K27me3 dysregulation and allelic skewing.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

Data availability

Access to data supporting this study is restricted due to the protection of participant confidentiality. Data can be accessed upon request with permission of the third party and approval of UW-Madison and Institutional Review Board.

References

  1. Collison J (2018) Autoimmunity: the ABCs of autoimmune disease. Nat Rev Rheumatol 14:248. https://doi.org/10.1038/nrrheum.2018.39

    Article  PubMed  Google Scholar 

  2. Whitacre CC (2001) Sex differences in autoimmune disease. Nat Immunol 2:777–780. https://doi.org/10.1038/ni0901-777

    Article  CAS  PubMed  Google Scholar 

  3. Manuel RSJ, Liang Y (2021) Sexual dimorphism in immunometabolism and autoimmunity: impact on personalized medicine. Autoimmun Rev: 102775. https://doi.org/10.1016/j.autrev.2021.102775

  4. Fish EN (2008) The X-files in immunity: sex-based differences predispose immune responses. Nat Rev Immunol 8:737–744. https://doi.org/10.1038/nri2394

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Liu K, Kurien BT, Zimmerman SL, Kaufman KM, Taft DH, Kottyan LC, Lazaro S, Weaver CA, Ice JA, Adler AJ et al (2016) X chromosome dose and sex bias in autoimmune diseases: increased prevalence of 47, XXX in systemic lupus erythematosus and Sjogren’s syndrome. Arthritis Rheumatol 68:1290–1300. https://doi.org/10.1002/art.39560

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Sharma R, Harris VM, Cavett J, Kurien BT, Liu K, Koelsch KA, Fayaaz A, Chaudhari KS, Radfar L, Lewis D et al (2017) Rare X chromosome abnormalities in systemic lupus erythematosus and Sjogren’s syndrome. Arthritis Rheumatol 69:2187–2192. https://doi.org/10.1002/art.40207

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Scofield RH, Bruner GR, Namjou B, Kimberly RP, Ramsey-Goldman R, Petri M, Reveille JD, Alarcon GS, Vila LM, Reid J et al (2008) Klinefelter’s syndrome (47, XXY) in male systemic lupus erythematosus patients: support for the notion of a gene-dose effect from the X chromosome. Arthritis Rheum 58:2511–2517. https://doi.org/10.1002/art.23701

    Article  PubMed  PubMed Central  Google Scholar 

  8. Augui S, Nora EP, Heard E (2011) Regulation of X-chromosome inactivation by the X-inactivation centre. Nat Rev Genet 12:429–442. https://doi.org/10.1038/nrg2987

    Article  CAS  PubMed  Google Scholar 

  9. Fang H, Disteche CM, Berletch JB (2019) X inactivation and escape: epigenetic and structural features. Front Cell Dev Biol 7. https://doi.org/10.3389/fcell.2019.00219

  10. Lyon MF (2000) LINE-1 elements and X chromosome inactivation: a function for “junk” DNA? Proc Natl Acad Sci U S A 97:6248–6249. https://doi.org/10.1073/pnas.97.12.6248

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Berletch JB, Yang F, Xu J, Carrel L, Disteche CM (2011) Genes that escape from X inactivation. Hum Genet 130:237–245. https://doi.org/10.1007/s00439-011-1011-z

    Article  PubMed  PubMed Central  Google Scholar 

  12. Ozbalkan Z, Bagislar S, Kiraz S, Akyerli CB, Ozer HT, Yavuz S, Birlik AM, Calguneri M, Ozcelik T (2005) Skewed X chromosome inactivation in blood cells of women with scleroderma. Arthritis Rheum 52:1564–1570. https://doi.org/10.1002/art.21026

    Article  CAS  PubMed  Google Scholar 

  13. Ozcelik T, Uz E, Akyerli CB, Bagislar S, Mustafa CA, Gursoy A, Akarsu N, Toruner G, Kamel N, Gullu S (2006) Evidence from autoimmune thyroiditis of skewed X-chromosome inactivation in female predisposition to autoimmunity. Eur J Hum Genet 14:791–797. https://doi.org/10.1038/sj.ejhg.5201614

    Article  CAS  PubMed  Google Scholar 

  14. Brix TH, Knudsen GP, Kristiansen M, Kyvik KO, Orstavik KH, Hegedus L (2005) High frequency of skewed X-chromosome inactivation in females with autoimmune thyroid disease: a possible explanation for the female predisposition to thyroid autoimmunity. J Clin Endocrinol Metab 90:5949–5953. https://doi.org/10.1210/jc.2005-1366

    Article  CAS  PubMed  Google Scholar 

  15. Brandt JE, Priori R, Valesini G, Fairweather D (2015) Sex differences in Sjögren’s syndrome: a comprehensive review of immune mechanisms. Biol Sex Differ 6:19–19

    Article  Google Scholar 

  16. Mougeot J-L, Noll B, Bahrani Mougeot F (2019) Sjögren’s syndrome X-chromosome dose effect: An epigenetic perspective. Oral Dis 25: 372-384. https://doi.org/10.1111/odi.12825

  17. McCoy SS, Giri J, Das R, Paul PK, Pennati A, Parker M, Liang Y, Galipeau J (2021) Minor salivary gland mesenchymal stromal cells derived from patients with Sjgren’s syndrome deploy intact immune plasticity. Cytotherapy 23:301–310. https://doi.org/10.1016/j.jcyt.2020.09.008

    Article  CAS  PubMed  Google Scholar 

  18. Yoo C, Vines JB, Alexander G, Murdock K, Hwang P, Jun HW (2014) Adult stem cells and tissue engineering strategies for salivary gland regeneration: a review. Biomater Res 18:9. https://doi.org/10.1186/2055-7124-18-9

    Article  PubMed  PubMed Central  Google Scholar 

  19. Iyer SS, Rojas M (2008) Anti-inflammatory effects of mesenchymal stem cells: novel concept for future therapies. Expert Opin Biol Ther 8:569–581. https://doi.org/10.1517/14712598.8.5.569

    Article  CAS  PubMed  Google Scholar 

  20. Choi EW (2009) Adult stem cell therapy for autoimmune disease. Int J Stem Cells 2:122–128. https://doi.org/10.15283/ijsc.2009.2.2.122

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Jalili V, Afgan E, Gu Q, Clements D, Blankenberg D, Goecks J, Taylor J, Nekrutenko A (2020) The Galaxy platform for accessible, reproducible and collaborative biomedical analyses: 2020 update. Nucleic Acids Res 48:W395–W402. https://doi.org/10.1093/nar/gkaa434

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Daniels TE, Cox D, Shiboski CH, Schiodt M, Wu A, Lanfranchi H, Umehara H, Zhao Y, Challacombe S, Lam MY et al (2011) Associations between salivary gland histopathologic diagnoses and phenotypic features of Sjogren’s syndrome among 1,726 registry participants. Arthritis Rheum 63:2021–2030. https://doi.org/10.1002/art.30381

    Article  PubMed  PubMed Central  Google Scholar 

  23. Castel SE, Levy-Moonshine A, Mohammadi P, Banks E, Lappalainen T (2015) Tools and best practices for data processing in allelic expression analysis. Genome Biol 16:195. https://doi.org/10.1186/s13059-015-0762-6

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Rubtsova K, Marrack P, Rubtsov AV (2015) Sexual dimorphism in autoimmunity. J Clin Invest 125:2187–2193. https://doi.org/10.1172/JCI78082

    Article  PubMed  PubMed Central  Google Scholar 

  25. Libert C, Dejager L, Pinheiro I (2010) The X chromosome in immune functions: when a chromosome makes the difference. Nat Rev Immunol 10:594–604. https://doi.org/10.1038/nri2815

    Article  CAS  PubMed  Google Scholar 

  26. Christou EAA, Banos A, Kosmara D, Bertsias GK, Boumpas DT (2019) Sexual dimorphism in SLE: above and beyond sex hormones. Lupus 28:3–10. https://doi.org/10.1177/0961203318815768

    Article  CAS  PubMed  Google Scholar 

  27. Mougeot JL, Noll BD, Bahrani Mougeot FK (2019) Sjogren’s syndrome X-chromosome dose effect: An epigenetic perspective. Oral Dis 25:372–384. https://doi.org/10.1111/odi.12825

    Article  PubMed  Google Scholar 

  28. Sachidanandam R, Weissman D, Schmidt SC, Kakol JM, Stein LD, Marth G, Sherry S, Mullikin JC, Mortimore BJ, Willey DL et al (2001) A map of human genome sequence variation containing 1.42 million single nucleotide polymorphisms. Nature 409:928–933. https://doi.org/10.1038/35057149

    Article  CAS  PubMed  Google Scholar 

  29. Ma S, Xie N, Li W, Yuan B, Shi Y, Wang Y (2014) Immunobiology of mesenchymal stem cells. Cell Death Differ 21:216–225. https://doi.org/10.1038/cdd.2013.158

    Article  CAS  PubMed  Google Scholar 

  30. Balaton BP, Cotton AM, Brown CJ (2015) Derivation of consensus inactivation status for X-linked genes from genome-wide studies. Biol Sex Differ 6:35. https://doi.org/10.1186/s13293-015-0053-7

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Tukiainen T, Villani AC, Yen A, Rivas MA, Marshall JL, Satija R, Aguirre M, Gauthier L, Fleharty M, Kirby A et al (2017) Landscape of X chromosome inactivation across human tissues. Nature 550:244–248. https://doi.org/10.1038/nature24265

    Article  PubMed  PubMed Central  Google Scholar 

  32. Gallinari P, Di Marco S, Jones P, Pallaoro M, Steinkuhler C (2007) HDACs, histone deacetylation and gene transcription: from molecular biology to cancer therapeutics. Cell Res 17:195–211. https://doi.org/10.1038/sj.cr.7310149

    Article  CAS  PubMed  Google Scholar 

  33. Thul PJ, Akesson L, Wiking M, Mahdessian D, Geladaki A, Ait Blal H, Alm T, Asplund A, Bjork L, Breckels LM et al (2017) A subcellular map of the human proteome. Science 356. https://doi.org/10.1126/science.aal3321

  34. Chow JC, Ciaudo C, Fazzari MJ, Mise N, Servant N, Glass JL, Attreed M, Avner P, Wutz A, Barillot E et al (2016) LINE-1 activity in facultative heterochromatin formation during X chromosome inactivation. Cell 166:782. https://doi.org/10.1016/j.cell.2016.07.013

    Article  CAS  PubMed  Google Scholar 

  35. Nezos A, Mavragani CP (2015) Contribution of genetic factors to Sjogren’s syndrome and Sjogren’s syndrome related lymphomagenesis. J Immunol Res 2015:754825. https://doi.org/10.1155/2015/754825

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Teos LY, Alevizos I (2017) Genetics of Sjogren’s syndrome. Clin Immunol 182:41–47. https://doi.org/10.1016/j.clim.2017.04.018

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Gershwin ME, Terasaki I, Graw R, Chused TM (1975) Increased frequency of HL-A8 in Sjogren’s syndrome. Tissue Antigens 6:342–346. https://doi.org/10.1111/j.1399-0039.1975.tb00653.x

    Article  CAS  PubMed  Google Scholar 

  38. Kim S, Yu NK, Kaang BK (2015) CTCF as a multifunctional protein in genome regulation and gene expression. Exp Mol Med 47:e166. https://doi.org/10.1038/emm.2015.33

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Pelekanos RA, Li J, Gongora M, Chandrakanthan V, Scown J, Suhaimi N, Brooke G, Christensen ME, Doan T, Rice AM et al (2012) Comprehensive transcriptome and immunophenotype analysis of renal and cardiac MSC-like populations supports strong congruence with bone marrow MSC despite maintenance of distinct identities. Stem Cell Res 8:58–73. https://doi.org/10.1016/j.scr.2011.08.003

    Article  CAS  PubMed  Google Scholar 

  40. Shockley KR, Lazarenko OP, Czernik PJ, Rosen CJ, Churchill GA, Lecka-Czernik B (2009) PPARgamma2 nuclear receptor controls multiple regulatory pathways of osteoblast differentiation from marrow mesenchymal stem cells. J Cell Biochem 106:232–246. https://doi.org/10.1002/jcb.21994

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Chougule A, Kolli V, Baroi S, Ebraheim N, Czernik PJ, Loh YP, Lecka-Czernik B (2020) Nonenzymatic and trophic activities of carboxypeptidase e regulate bone mass and bioenergetics of skeletal stem cells in mice. JBMR Plus 4:e10392. https://doi.org/10.1002/jbm4.10392

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Wang Y, Tong X, Omoregie ES, Liu W, Meng S, Ye X (2012) Tetraspanin 6 (TSPAN6) negatively regulates retinoic acid-inducible gene I-like receptor-mediated immune signaling in a ubiquitination-dependent manner. J Biol Chem 287:34626–34634. https://doi.org/10.1074/jbc.M112.390401

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ruffell B, Poon GF, Lee SS, Brown KL, Tjew SL, Cooper J, Johnson P (2011) Differential use of chondroitin sulfate to regulate hyaluronan binding by receptor CD44 in inflammatory and interleukin 4-activated macrophages. J Biol Chem 286:19179–19190. https://doi.org/10.1074/jbc.M110.200790

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Galupa R, Heard E (2018) X-chromosome inactivation: a crossroads between chromosome architecture and gene regulation. Annu Rev Genet 52:535–566. https://doi.org/10.1146/annurev-genet-120116-024611

    Article  CAS  PubMed  Google Scholar 

  45. Brooks WH, Renaudineau Y (2015) Epigenetics and autoimmune diseases: the X chromosome-nucleolus nexus. Front Genet 6:22. https://doi.org/10.3389/fgene.2015.00022

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Gough SC, Simmonds MJ (2007) The HLA region and autoimmune disease: associations and mechanisms of action. Curr Genomics 8:453–465. https://doi.org/10.2174/138920207783591690

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Chitnis N, Clark PM, Kamoun M, Stolle C, Brad Johnson F, Monos DS (2017) An Expanded Role for HLA Genes: HLA-B encodes a microRNA that regulates IgA and other immune response transcripts. Front Immunol 8:583. https://doi.org/10.3389/fimmu.2017.00583

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Zhang J, Zhang YZ, Jiang J, Duan CG (2020) The crosstalk between epigenetic mechanisms and alternative RNA processing regulation. Front Genet 11:998. https://doi.org/10.3389/fgene.2020.00998

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Zito A, Davies MN, Tsai PC, Roberts S, Andres-Ejarque R, Nardone S, Bell JT, Wong CCY, Small KS (2019) Heritability of skewed X-inactivation in female twins is tissue-specific and associated with age. Nat Commun 10:5339. https://doi.org/10.1038/s41467-019-13340-w

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Funding

SSM has received research support from the Clinical and Translational Science Award through the NIH National Center for Advancing Translational Sciences, grant UL1TR002373 and KL2TR002374. JG is funded by the NIH National Institute of Diabetes and Digestive and Kidney Diseases award R01 DK109508. YL has received research support from the NIH National Institute of Arthritis and Musculoskeletal and Skin Diseases grant K01 AR073340 and Wisconsin Partnership Program New Investigator Program.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yun Liang.

Ethics declarations

Ethics approval

This study was performed in line with the principles of the Declaration of Helsinki. Approval of human studies was granted by the UW-Madison Institutional Review Board.

Consent to participate

Informed consent was obtained from all individual participants included in the study.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary Table. Table of skewed X-linked genes in pSS.

X-linked genes skewed in each pSS subject are listed with gene symbol, Ensemble ID and minor allele frequency (MAF). (XLSX 18 KB)

Supplementary file2 (DOCX 41 KB)

Supplementary Fig. 1. Demographic and clinical profiles of control and pSS subjects.

Demographic and clinical information of control and pSS subjects are shown, including age, sex, race, group, SSA status, focus score, ANA, rheumatoid factor, C3, C4, organ involvement, risk factor for lymphoma, children, postmenopausal, contraceptive, hormone replacement, and surgical menopause. Supplementary Fig. 2. Venn Diagram of skewed, X-linked genes in pSS subjects. The number of overlapping and non-overlapping skewed, X-linked genes in the four pSS subjects (SS1, SS2, SS3 and SS4) are shown by the Venn Diagram. Supplementary Fig. 3. Stability of allele-specific expression and DNA allelic ratio of skewed expressors. (a) Allele-specific qPCR of TSPAN6 transcripts in control (N) and pSS (SS), passage 1-4 MSCs (n=4 independent subjects each), showing stability in allele-specific expression. Similar (i.e., non-drifting) results have been obtained for CHST7, ZDHHC9 and BCOR. (b) DNA allelic ratio of TSPAN6, CHST7 and ZDHCC9 in control (N) and pSS (SS) MSCs by allele-specific qPCR of DNA, showing ~1:1 ratio of the two alleles with no evidence of aneuploidy. Data shown were from n=3 replicates from one subject, and result was confirmed in samples from an independent subject with skewed expression of given target. Mean ± sem. Supplementary Fig. 4. Allele-specific expression analysis of XIST. Allele-specific qPCR of XIST in control (N) and pSS (SS) MSCs, showing lack of skewing in at least two of the four pSS MSCs. Supplementary Fig. 5. Skewing in whole minor salivary glands in pSS and in blood and skin of SLE. (a) Allele-specific qPCR for representative skewed expressors (TSPAN6, CHST7, ZDHHC9) and BCOR as a control, showing skewed expression of X-linked genes in pSS but not control whole salivary glands (n=4 pSS, n=4 control, independent subjects each, female). N, control. SS, pSS. WG, whole minor salivary gland. (b) Allele-specific qPCR for TSPAN6, CHST7, ZDHHC9 and BCOR, showing skewing of TSPAN6 and BCOR in SLE but not control blood cells (n=6 SLE, n=5 control, independent subjects each, female). N, control. SLE, systemic lupus erythematosus. PBMC, peripheral blood mononuclear cells. (c) Allele-specific qPCR for TSPAN6, CHST7, ZDHHC9 and BCOR, showing skewing of TSPAN6 and BCOR in SLE but not control skin (n=5 SLE, n=2 control, independent subjects each, female). N, control. SLE, systemic lupus erythematosus. Supplementary Fig. 6. Allele-specific expression of select genes on the X chromosome. (a) Allele-specific expression of immune-associated genes in each control (N) or pSS (SS) sample with skewed expression indicated in yellow (MAF < 0.35), showing lack of pSS-associated skewing for these genes. Grey boxes show missing data (no detected common het-SNPs) from RNA-Seq. (b) Allele-specific expression of common variants in each control (N) or pSS (SS) sample with skewed expression indicated in yellow (MAF < 0.35), showing pSS-associated skewing. Grey boxes show missing data (no detected common het-SNPs) from RNA-Seq. (c) Gene length for genes listed in (A, immune-associated) or (B, genes with common variants), showing no significant difference in length between the two groups. (d) Total RNA expression levels for genes listed in (A, immune-associated) or (B, genes with common variants) in control (N) or pSS (SS) MSCs, showing no significant difference in RNA expression levels between the two groups. Supplementary Fig. 7. Skewed expressors associate with histone modification and chromatin organization functions. (a) Network analysis of skewed expressors, showing the three biological networks formed by these genes. (b) Biological process enrichment analysis, showing histone H2A acetylation, protein acylation and chromatin organization as the top biological processes enriched in skewed expressors (FDR < 0.05). (c) Cellular component analysis of skewed expressors, showing enrichment of the NuA4 histone acetylatransferase complex, Swr1 complex and histone deacetylase complex (FDR < 0.05). Supplementary Fig. 8. Protein descriptions for the skewed expressor network. Proteins in the three biological networks formed by skewed expressors, as shown in Supplementary Fig. 5, are described with cluster information, protein name and functional description. Supplementary Fig. 9. Expression levels of skewed expressors in pSS and control MSCs. (a) qPCR of indicated genes (APOO, CHST7, PJA1, TSPAN6, TCEAL4, MORF4L2, SLC25A43, GRIA3, ZDHCC9, AIFM1, MOSPD1, LDOC1, IDS, FLNA) from pSS (SS) and control (N) MSCs, showing comparable expression levels in control and pSS MSCs (n=4 independent subjects each). Mean ± sem. (b) Western blot analysis of indicated proteins (TSPAN6, CHST7 and control GAPDH) in pSS (SS) and control (N) MSCs (4-20% gradient gel, loading equal amounts of protein amounts; markers indicated on gel), showing comparable expression levels in control and pSS MSCs. Supplementary Fig. 10. BCOR localization in MSCs. Immunostaining of BCOR and DAPI in control (N) and pSS (SS) MSCs, showing comparable localization in the two groups (images representative of > 100 cells in four independent subjects each). Supplementary Fig. 11. Copy number analysis in control and pSS MSCs. DNA copy number analysis of genes on the X chromosome and autosomes (chromosome 12 and chromosome 2), showing lack of altered ploidy in pSS MSCs (SS) compared to control (N) (n=4 independent subjects each). Mean ± sem. Supplementary Fig. 12. XIST decrease leads to disease-associated mislocalization of skewed expressors. (a) qPCR of XIST and LINE1 (L1) levels upon scrambled (scr siRNA) or XIST knockdown (XIST siRNA) in control MSCs, showing decrease in XIST upon XIST knockdown. Data shown were from n=3 replicates from one subject, and result was confirmed in samples from two independent subjects. Mean ± sem, * P < 0.05, Student’s t-test, two tailed. Line under asterisk indicates groups compared. (b) Immunostaining of TSPAN6 and DAPI in control MSCs with scrambled (scr siRNA) or XIST knockdown (XIST siRNA), showing ectopic TSPAN6+ foci upon XIST knockdown (pointed by arrow; images representative of > 100 cells from three independent subjects). (c) Immunostaining of CHST7 and DAPI in control MSCs with scrambled (scr siRNA) or XIST knockdown (XIST siRNA), showing loss of CHST7+ foci upon XIST knockdown (pointed by arrow; > 100 cells from three independent subjects). Supplementary Fig. 13. Knockdown efficiency of CTCF siRNA and specificity of miR6891-5p inhibition. (a) qPCR of CTCF levels (left two bars) and miR-3135B levels (right two bars) upon control or miR6891-5p inhibition (n=4 independent subjects). (b) qPCR of CTCF levels upon scrambled or CTCF knockdown in control MSCs, showing decrease in CTCF upon CTCF knockdown (n=4 independent subjects). Mean ± sem, * P < 0.05, Student’s t-test, two tailed. Line under asterisk indicates groups compared. Supplementary Fig. 14. miR6891-5p inhibition leads to disease-associated mislocalization of skewed genes. (a) Immunostaining of TSPAN6 and DAPI in control or miR6891-5p-inhibited MSCs, showing ectopic TSPAN6+ foci upon miR6891-5p inhibition (pointed by arrow; > 100 cells from three independent subjects). (b) Immunostaining of CHST7 and DAPI in control or miR6891-5p-inhibited MSCs, showing loss of CHST7+ foci upon miR6891-5p inhibition (pointed by arrow; > 100 cells from three independent subjects). (c) Allele-specific ChIP of IgG, H3K27me3 and H3K36me3 on BCOR in MSCs with control (Scr siRNA), XIST knockdown (XIST siRNA) or miR6891-5p inhibition (miR6891-5p inh), showing that XIST knockdown or miR6891-5p inhibition does not alter epigenetic state of non-skewed expressors. Data shown were from n=3 replicates from one subject, and result was confirmed in samples from an independent subject. Mean ± sem. Supplementary Fig. 15. pSS MSCs exhibit inflammatory differentiation. (a) qPCR of IL6 in control (N) and pSS (SS) MSCs during differentiation, showing comparable expression levels of IL6 in control and pSS MSCs. (b) qPCR of IL4 in control (N) and pSS (SS) MSCs during differentiation, showing deficiency in IL4 upregulation upon pSS MSC differentiation. (c, d) qPCR of IL4 (c) and IL1B (d) in MSCs with control, XIST knockdown (XIST Ri) or miR6891 inhibition (miR6891-5p inh), showing deficiency in IL4 upregulation during pSS MSC differentiation upon XIST and miR6891 disruption. n=3 independent subjects each. Mean ± sem, * P < 0.05, Student’s t-test, two tailed. Line under asterisk indicates groups compared. (PDF 5174 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shaw, T.M., Zhang, W., McCoy, S.S. et al. X-linked genes exhibit miR6891-5p-regulated skewing in Sjögren’s syndrome. J Mol Med 100, 1253–1265 (2022). https://doi.org/10.1007/s00109-022-02205-3

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00109-022-02205-3

Keywords

Navigation