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.

  • Article
  • Published:

Nuclear smooth muscle α-actin participates in vascular smooth muscle cell differentiation

Abstract

Missense variants throughout ACTA2, encoding α-smooth muscle actin (αSMA), predispose to adult-onset thoracic aortic disease, but variants disrupting arginine 179 (R179) lead to smooth muscle dysfunction syndrome characterized by diverse childhood-onset vascular diseases. Here we show that αSMA localizes to the nucleus in wild-type smooth muscle cells (SMCs), enriches in the nucleus with SMC differentiation, and associates with chromatin remodeling complexes and SMC contractile gene promoters. The ACTA2 p.Arg179 αSMA variant shows decreased nuclear localization. Primary SMCs from Acta2SMC-R179C/+ mice are less differentiated than wild-type SMCs in vitro and in vivo and have global changes in chromatin accessibility. Induced pluripotent stem cells from participants with ACTA2 p.Arg179 variants fail to fully differentiate from neuroectodermal progenitor cells to SMCs, and single-cell transcriptomic analyses of an ACTA2 p.Arg179His participant’s aortic tissue show increased SMC plasticity. Thus, nuclear αSMA participates in SMC differentiation, and loss of this nuclear activity occurs with ACTA2 p.Arg179 pathogenic variants.

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

Access options

Buy this article

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

Fig. 1: α-smooth muscle actin localizes to the nucleus concurrently with smooth muscle cell differentiation.
Fig. 2: α-smooth muscle actin binds chromatin remodeling complexes and the promoters of smooth muscle cell contractile genes.
Fig. 3: p.Arg179Cys mutation impairs nuclear α-smooth muscle actin localization and function when expressed in Acta2−/− SMCs.
Fig. 4: Heterozygous inducible knock-in of the p.Arg179Cys mutation in mouse smooth muscle cells leads to loss of nuclear α-smooth muscle actin and decreased differentiation of smooth muscle cells.
Fig. 5: Acta2SMC-R179C/+ mouse smooth muscle cells have altered chromatin accessibility in vitro and are hypodifferentiated in vivo.
Fig. 6: Heterozygous participant-derived ACTA2 p.Arg179Cys smooth muscle cells are less differentiated with reduced nuclear αSMA.
Fig. 7: Single-cell RNA sequencing of ACTA2 p.Arg179His participant tissue confirms hypodifferentiated phenotype and increased plasticity in vivo.

Similar content being viewed by others

Data availability

scRNA-seq datasets generated for this manuscript are available in the GEO under accession number GSE201091. The ATAC-seq dataset generated for this manuscript has been deposited in the GEO and is available under accession number GSE241055. All reagents and resources applicable to this study are available from the corresponding authors upon reasonable request. Source data are provided with this paper.

Code availability

No novel code or algorithm was generated for this study.

References

  1. Guo, D. C. et al. Mutations in smooth muscle alpha-actin (ACTA2) cause coronary artery disease, stroke, and Moyamoya disease, along with thoracic aortic disease. Am. J. Hum. Genet. 84, 617–627 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Guo, D. C. et al. Mutations in smooth muscle alpha-actin (ACTA2) lead to thoracic aortic aneurysms and dissections. Nat. Genet. 39, 1488–1493 (2007).

    Article  CAS  PubMed  Google Scholar 

  3. Milewicz, D. M. et al. Altered smooth muscle cell force generation as a driver of thoracic aortic aneurysms and dissections. Arterioscler. Thromb. Vasc. Biol. 37, 26–34 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Milewicz, D. M. et al. De novo ACTA2 mutation causes a novel syndrome of multisystemic smooth muscle dysfunction. Am. J. Med. Genet. A 152A, 2437–2443 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Regalado, E. S. et al. Clinical history and management recommendations of the smooth muscle dysfunction syndrome due to ACTA2 arginine 179 alterations. Genet. Med. 20, 1206–1215 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Lauer, A. et al. Cerebrovascular disease progression in patients with ACTA2 Arg179 pathogenic variants. Neurology 96, e538–e552 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Munot, P. et al. A novel distinctive cerebrovascular phenotype is associated with heterozygous Arg179 ACTA2 mutations. Brain 135, 2506–2514 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Georgescu, M. M. et al. The defining pathology of the new clinical and histopathologic entity ACTA2-related cerebrovascular disease. Acta Neuropathol. Commun. 3, 81–87 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Kelpsch, D. J. & Tootle, T. L. Nuclear actin: from discovery to function. Anat. Rec. 301, 1999–2013 (2018).

    Article  CAS  Google Scholar 

  10. Xie, X. et al. β‐actin‐dependent global chromatin organization and gene expression programs control cellular identity. FASEB J. 32, 1296–1314 (2019).

    Google Scholar 

  11. Xie, X., Jankauskas, R., Mazari, A. M. A., Drou, N. & Percipalle, P. β-actin regulates a heterochromatin landscape essential for optimal induction of neuronal programs during direct reprograming. PLoS Genet. 14, e1007846 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Kumar, A. et al. Actin R256 mono-methylation is a conserved post-translational modification involved in transcription. Cell. Rep. 32, 108172 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ilik, I. A. & Aktas, T. Nuclear speckles: dynamic hubs of gene expression regulation. FEBS J. 289, 7234–7245 (2022).

    Article  CAS  PubMed  Google Scholar 

  14. Cheung, C., Bernardo, A. S., Pedersen, R. A. & Sinha, S. Directed differentiation of embryonic origin-specific vascular smooth muscle subtypes from human pluripotent stem cells. Nat. Protoc. 9, 929–938 (2014).

    Article  CAS  PubMed  Google Scholar 

  15. Owens, G. K., Kumar, M. S. & Wamhoff, B. R. Molecular regulation of vascular smooth muscle cell differentiation in development and disease. Physiol. Rev. 84, 767–801 (2004).

    Article  CAS  PubMed  Google Scholar 

  16. Kaw, A. et al. Mosaicism for the smooth muscle cell (SMC)-specific knock-in of the Acta2 R179C pathogenic variant: implications for gene editing therapies. J. Mol. Cell. Cardiol. 171, 102–104 (2022).

    Article  CAS  PubMed  Google Scholar 

  17. Liedtke, S., Stephan, M. & Kogler, G. Oct4 expression revisited: potential pitfalls for data misinterpretation in stem cell research. Biol. Chem. 389, 845–850 (2008).

    Article  CAS  PubMed  Google Scholar 

  18. Mayor, R. & Theveneau, E. The neural crest. Development 140, 2247–2251 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. Lu, H., Fagnant, P. M., Krementsova, E. B. & Trybus, K. M. Severe molecular defects exhibited by the R179H mutation in human vascular smooth muscle alpha-actin. J. Biol. Chem. 291, 21729–21739 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hinz, B., Gabbiani, G. & Chaponnier, C. The NH2-terminal peptide of alpha-smooth muscle actin inhibits force generation by the myofibroblast in vitro and in vivo. J. Cell Biol. 157, 657–663 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Papke, C. L. et al. Smooth muscle hyperplasia due to loss of smooth muscle alpha-actin is driven by activation of focal adhesion kinase, altered p53 localization and increased levels of platelet-derived growth factor receptor-beta. Hum. Mol. Genet. 22, 3123–3137 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yap, C., Mieremet, A., de Vries, C. J. M., Micha, D. & de Waard, V. Six shades of vascular smooth muscle cells illuminated by klf4 (krüppel-like factor 4). Arter. Thromb. Vasc. Biol. 41, 2693–2707 (2021).

    Article  CAS  Google Scholar 

  23. Kato, S. et al. Ectopic expression of Smad7 inhibits transforming growth factor-β responses in vascular smooth muscle cells. Life Sci. 69, 2641–2652 (2001).

    Article  PubMed  Google Scholar 

  24. Hideto, O. et al. Smooth muscle cell phenotype-dependent transcriptional regulation of the alpha-1 integrin gene. J. Biol. Chem. 272, 26643–26651 (1997).

    Article  Google Scholar 

  25. Turczyńska, K. M. et al. Regulation of smooth muscle dystrophin and synaptopodin 2 expression by actin polymerization and vascular Injury. Arter. Thromb. Vasc. Biol. 35, 1489–1497 (2015).

    Article  Google Scholar 

  26. Chen, J. et al. Loss of smooth muscle alpha-actin leads to NF-κB-dependent increased sensitivity to angiotensin II in smooth muscle cells and aortic enlargement. Circ. Res. 120, 1903–1915 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Schildmeyer, L. A. et al. Impaired vascular contractility and blood pressure homeostasis in the smooth muscle alpha-actin null mouse. FASEB J. 14, 2213–2220 (2000).

    Article  CAS  PubMed  Google Scholar 

  28. Pedroza, A. J. et al. Single-cell transcriptomic profiling of vascular smooth muscle cell phenotype modulation in Marfan syndrome aortic aneurysm. Arterioscler. Thromb. Vasc. Biol. 40, 2195–2211 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Wirka, R. C. et al. Atheroprotective roles of smooth muscle cell phenotypic modulation and the TCF21 disease gene as revealed by single-cell analysis. Nat. Med. 25, 1280–1289 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Schwarz, D. et al. Ezh2 is required for neural crest-derived cartilage and bone formation. Development 141, 867–877 (2014).

    Article  CAS  PubMed  Google Scholar 

  33. Takizawa, H. et al. Neural crest-derived cells possess differentiation potential to keratinocytes in the process of wound healing. Biomed. Pharmacother. 146, 112593 (2022).

    Article  CAS  PubMed  Google Scholar 

  34. Crane, J. F. & Trainor, P. A. Neural crest stem and progenitor cells. Annu. Rev. Cell Dev. Biol. 22, 267–286 (2006).

    Article  CAS  PubMed  Google Scholar 

  35. Pan, H. et al. Single-cell genomics reveals a novel cell state during smooth muscle cell phenotypic switching and potential therapeutic targets for atherosclerosis in mouse and human. Circulation 142, 2060–2075 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Li, Y. et al. Single-cell transcriptome analysis reveals dynamic cell populations and differential gene expression patterns in control and aneurysmal human aortic tissue. Circulation 142, 1374–1388 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Mahmood, S. R. et al. β-actin dependent chromatin remodeling mediates compartment level changes in 3D genome architecture. Nat. Commun. 12, 5240 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Su, I. et al. Polycomb group protein ezh2 controls actin polymerization and cell signaling. Cell 121, 425–436 (2005).

    Article  CAS  PubMed  Google Scholar 

  39. Gunasekaran, S., Miyagawa, Y. & Miyamoto, K. Actin nucleoskeleton in embryonic development and cellular differentiation. Curr. Opin. Cell Biol. 76, 102100 (2022).

    Article  CAS  PubMed  Google Scholar 

  40. Lino Cardenas, C. L., Briere, L. C., Sweetser, D. A., Lindsay, M. E. & Musolino, P. L. A seed sequence variant in miR-145-5p causes multisystem smooth muscle dysfunction syndrome. J. Clin. Invest. 133, e166497 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Wang, Y. et al. Endogenous miRNA sponge lincRNA-RoR regulates Oct4, Nanog, and Sox2 in human embryonic stem cell self-renewal. Dev. Cell 25, 69–80 (2013).

    Article  CAS  PubMed  Google Scholar 

  42. Cordes, K. R. et al. miR-145 and miR-143 regulate smooth muscle cell fate and plasticity. Nature 460, 705–710 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Guo, D. C. et al. Loss-of-function mutations in YY1AP1 lead to Grange syndrome and a fibromuscular dysplasia-like vascular disease. Am. J. Hum. Genet. 100, 21–30 (2017).

    Article  CAS  PubMed  Google Scholar 

  44. Zhang, M., Fang, H., Zhou, J. & Herring, B. P. A novel role of Brg1 in the regulation of SRF/MRTFA-dependent smooth muscle-specific gene expression. J. Biol. Chem. 282, 25708–25716 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Zhou, J. et al. The SWI/SNF chromatin remodeling complex regulates myocardin-induced smooth muscle-specific gene expression. Arterioscler. Thromb. Vasc. Biol. 29, 921–928 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Bohnsack, M. T., Stuven, T., Kuhn, C., Cordes, V. C. & Gorlich, D. A selective block of nuclear actin export stabilizes the giant nuclei of Xenopus oocytes. Nat. Cell Biol. 8, 257–263 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Le, H. Q. et al. Mechanical regulation of transcription controls Polycomb-mediated gene silencing during lineage commitment. Nat. Cell Biol. 18, 864–875 (2016).

    Article  CAS  PubMed  Google Scholar 

  48. Neely, A. & Bao, X. Nuclei Isolation Staining (NIS) method for imaging chromatin-associated proteins in difficult cell types. Curr. Protoc. Cell Biol. 84, e94 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Cheung, C. et al. Generation of human vascular smooth muscle subtypes provides insight into embryological origin–dependent disease susceptibility. Nat. Biotechnol. 30, 165–173 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kwartler, C. S. et al. Vascular smooth muscle cell isolation and culture from mouse aorta. Bio Protoc. 6, e2045 (2016).

    Google Scholar 

  51. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by an America Heart Association Merit Award (to D.M.M.), the National Heart, Lung and Blood Institute (RO1 HL146583 to D.M.M.), the Olivia Petrera-Cohen Research Fund (to D.M.M.), the John Ritter Foundation (to D.M.M.), Marylin and Frederick R. Lummis, MD, Fellowship in the Biomedical Sciences (to A.K.), National Institutes of Health (NIH) TL1TR003169 (to A.K.), NIH UL1TR003167 (to A.K.), NIH F32HL154681 (to A.J.P.), NIH R01HL157949 (to M.P.F.), British Heart Foundation awards RG/17/5/32936 and FS/18/46/33663 (to S.S.) and American Heart Association grant 20CDA35310689 (to C.S.K.). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. Confocal microscopy was performed at the Center for Advanced Microscopy, Department of Integrative Biology and Pharmacology at McGovern Medical School, UTHealth. We thank G. Gabbiani and C. Chaponnier from the University of Geneva for their generosity in sharing the SKAfp and SMAfp peptides for this study.

Author information

Authors and Affiliations

Authors

Contributions

D.M.M. and C.S.K. designed the study. C.S.K. planned the individual experiments. C.S.K., A.K., S.M., X.y.D., C.K., J.E.E.P., M.S.B.B. and J.C. performed the cellular experiments. A.K. and A.J.P. obtained the sample and analyzed the scRNA-seq on mouse tissue. A.J.P. and M.P.F. obtained the sample and analyzed the scRNA-seq on participant tissue. C.S.K. and A.J.P. obtained the sample and analyzed the ATAC-seq on cultured SMCs. P.G. performed the integrated analysis combining scRNA-seq and ATAC-seq datasets. Y.Z. and X.S. consulted on nuclear actin functions and contributed to the design of experiments. S.S. reprogrammed a participant stem cell line and assisted with the stem cell differentiation protocol. D.M.M. and C.S.K. interpreted the data and drafted the manuscript. D.M.M. and M.P.F. obtained funding for this work.

Corresponding authors

Correspondence to Callie S. Kwartler or Dianna M. Milewicz.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cardiovascular Research thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: Vesna Todorovic, in collaboration with the Nature Cardiovascular Research Team.

Additional information

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

Extended data

Extended Data Fig. 1 αSMA localizes to the nucleus concurrently with SMC differentiation.

A) Quantitation of immunoblot in Fig. 1A shows that TGFβ1 stimulation for 48 h increases protein levels of αSMA in both the nucleus and cytosol, while PDGF-BB treatment for 24 h does not affect αSMA nuclear localization (n=3 independent experiments). Significance was assessed by one-way ANOVA followed by Tukey’s multiple comparisons test. B) Quantitation of the latrunculin (LtA) treatment immunoblot shown in Fig. 1C shows that depolymerization of actins by LtA treatment does not affect nuclear localization of actin, supporting that the observed nuclear localization is accurate (n = 4 independent experiments). Significance was assessed by Kruskal-Wallis test with Dunn’s multiple comparisons test. C) Quantitation of the NEPC to SMC differentiation timecourse immunoblot shown in Fig. 1G shows that αSMA levels in both the nucleus and cytosol increase around 15-20 fold during the differentiation. Over the same timecourse, β-actin levels do not dramatically change, and nuclear accumulation is lower than cytosolic (n = 4 independent experiments using 2 different control cell lines). All data are presented as mean ± standard deviation.

Source data

Extended Data Fig. 2 Forced nuclear localization of actins does not affect Mkl1 levels or localization.

A,B) Immunostaining of NLS-infected SMCs with Mkl1 antibody (A) shows no change in Mkl1 localization or intensity with α-NLS or β-NLS infection, quantified in (B). n = 14 cells infected with empty vector (EV), 15 cells infected with β-NLS, and 11 cells infected with α-NLS across three independent staining experiments. Significance was assessed by one-way ANOVA with Tukey’s multiple comparisons test. All data are presented as mean ± standard deviation.

Source data

Extended Data Fig. 3 Mouse Acta2SMC-R179C/+ smooth muscle cells have decreased nuclear αSMA and are hypodifferentiated.

A) Quantitation of immunoblot in Fig. 4E shows significant and reproducible decreases in accumulation of contractile proteins in the Acta2SMC-R179C/+ cells compared with WT (n = 4 independent experiments). Significance was assessed by two-way ANOVA followed by Tukey’s multiple comparisons test. B) Quantitation of immunoblot in Fig. 4F shows decreased nuclear accumulation, of αSMA in the Acta2SMC-R179C/+ cells compared with WT. β-actin accumulation to the nucleus was variable across experiments (n = 4 independent experiments). Significance was assessed by two-way ANOVA followed by Tukey’s multiple comparisons test. C) Quantitation of immunoblot in Fig. 4G shows that LtA treatment further does moderately decrease nuclear accumulation of β-actin in Acta2SMC-R179C/+ cells but does not significantly affect αSMA nuclear accumulation (n = 3 independent experiments). Significance was assessed by two-way ANOVA followed by Tukey’s multiple comparisons test. D) Co-immunoprecipitation with Brg1 antibody confirms decreased association of both αSMA and β-actin with the Baf chromatin remodeling complex in in Acta2SMC-R179C/+ SMCs which is partially rescued by TGFβ1 treatment (n = 3 independent experiments). All data are presented as mean ± standard deviation.

Source data

Extended Data Fig. 4 Differential chromatin accessibility aligns with transcriptional changes in Acta2SMC-R179C/+ SMCs.

A) Distribution of chromatin accessibility peaks around the transcription start sites of nearby genes for peaks with increased accessibility in WT SMCs. B) Distribution of chromatin accessibility peaks around the transcription start sites of nearby genes for peaks with increased accessibility in Acta2SMC-R179C/+ SMCs. C) Visualization of integrated scRNA-seq and ATAC-seq datasets plots the log2FC in gene expression in vivo on the y-axis and the log2FC in chromatin accessibility in vitro on the x-axis. Genes indicated by dots inside the red circle have decreased accessibility and decreased expression in Acta2SMC-R179C/+ SMCs and were analyzed by GO enrichment analysis in Fig. 5H. Genes indicated by dots inside the blue circle have increased accessibility and increased expression in Acta2SMC-R179C/+ SMCs and were analyzed by GO enrichment analysis in Fig. 5H.

Extended Data Fig. 5 Genes with increased expression and accessibility in Acta2SMC-R179C/+ SMCs.

A) Genome browser view for the Klf4 gene shows chromatin accessibility increased in Acta2SMC-R179C/+ SMCs in vitro and transcript levels visualized in UMAP space and quantified shows increased expression in Acta2SMC-R179C/+ SMCs in vivo. B) Genome browser view for the Smad7 gene shows chromatin accessibility increased in Acta2SMC-R179C/+ SMCs in vitro and transcript levels visualized in UMAP space and quantified shows increased expression in Acta2SMC-R179C/+ SMCs in vivo. P values denote results of Wilcoxon rank sum test with Bonferroni correction for multiple comparisons between all ‘SMC 1’ (5,816 cells) and ‘SMC 2’ (3,229 cells) in the scRNAseq dataset. For all violin plots, line depicts median, hinges depict interquartile range, whiskers depict 1.5 interquartile ranges above and below the hinges. C) Quantitative RT-PCR validates shRNA-mediated knockdown of Smad7 in WT SMCs and shows knockdown of Smad7 increases expression of SMC contractile genes. Graph shows three technical replicates, representative of three independent experiments from three distinct lentiviral infections. Significance was assessed by unpaired, two-tailed t-test for each gene. All data are presented as mean ± standard deviation.

Source data

Extended Data Fig. 6 Genes with decreased expression and accessibility in Acta2SMC-R179C/+ SMCs.

A) Genome browser view for the Itga1 gene shows chromatin accessibility decreased in Acta2SMC-R179C/+ SMCs in vitro and transcript levels visualized in UMAP space and quantified shows decreased expression in Acta2SMC-R179C/+ SMCs in vivo. B) Genome browser view for the Synpo2 gene shows chromatin accessibility decreased in Acta2SMC-R179C/+ SMCs in vitro and transcript levels visualized in UMAP space and quantified shows decreased expression in Acta2SMC-R179C/+ SMCs in vivo. P values denote results of Wilcoxon rank sum test with Bonferroni correction for multiple comparisons between all ‘SMC 1’ (5,816 cells) and ‘SMC 2’ (3,229 cells) in the scRNAseq dataset. For all violin plots, line depicts median, hinges depict interquartile range, whiskers depict 1.5 interquartile ranges above and below the hinges.

Extended Data Fig. 7 Participant-derived ACTA2 p.Arg179 SMCs are less differentiated than controls.

A) Demographic and genotype information for patient and control lines used in this study. B) Quantitation of immunoblot from Fig. 6A reveals significant decreases in contractile protein accumulation in ACTA2 p.R179C patient SMCs compared with control SMCs (n = 4 independent experiments). Significance was assessed by one-way ANOVA with Sidak’s multiple comparisons test. C-E) Immunoblot showing decreased contractile protein accumulation is a consistent finding across three ACTA2 p.R179 patient lines (C), quantified in (D,E). Graph shows biological replicates- each dot represents one patient or control line. Significance in (D) ( + /- TGFβ1) was assessed by two-way ANOVA with Tukey’s multiple comparisons test. Significance in (E) (no TGFβ1) was assessed by unpaired, two-tailed t-tests per protein. Grey bars/black dots represent control and green bars/dots represent ACTA2 p.R179. All data are presented as mean ± standard deviation.

Source data

Extended Data Fig. 8 Participant-derived ACTA2 p.Arg179 SMCs have decreased nuclear αSMA compared with controls.

A) Quantitation of immunoblot from Fig. 6C shows a significant decrease in αSMA nuclear accumulation in patient-derived ACTA2 p.R179C SMCs compared with controls. β-actin accumulation to the nucleus was variable and not significantly altered (n = 3 independent experiments). Significance was assessed by repeated measures one-way ANOVA followed by Tukey’s multiple comparisons test. B,C) Immunoblot showing decreased nuclear accumulation of αSMA is a consistent phenotype across three ACTA2 p.R179 patient lines (B), quantified in (C). Graph shows biological replicates- each dot represents one patient or control line. Significance was assessed by unpaired, two-tailed t-test for each condition (cytosol, nucleus) for each protein. D) Quantitation of immunoblot from Fig. 6D shows a significant increase in cytosolic αSMA and decrease in αSMA nuclear accumulation in patient-derived ACTA2 p.R179 NEPCs compared with controls. β-actin accumulation to the nucleus was not significantly altered (n = 3 independent experiments). Significance was assessed by unpaired, two-tailed t-test for each condition (cytosol, nucleus) for each protein. E) Co-immunoprecipitation with BRG1 antibody confirms decreased association of both αSMA and β-actin with the BAF chromatin remodeling complex in ACTA2 p.R179C SMCs. Grey bars/black dots represent control and green bars/dots represent ACTA2 p.R179. All data are presented as mean ± standard deviation.

Source data

Extended Data Fig. 9 Expression of skeletal α-actin can compensate for loss of nuclear αSMA in Acta2-/- cells.

A) Immunostaining for αSMA shows partial disruption of αSMA filaments in SMCs with the ACTA2 p.R179C pathogenic variant. B) Subcellular fractionation immunoblot of Acta2 KO mouse SMCs shows that sarcomeric actin (encoded by ACTA1 and known to be expressed in Acta2 KO SMCs as a compensatory mechanism for loss of αSMA) in these cells can go into the nucleus. C) Quantitative RT-PCR confirms that ACTA2 Crispr-edited iPSC-derived SMCs do not have increased expression of ACTA1. Graph shows three technical replicates, results are representative of three independent experiments. All data are presented as mean ± standard deviation.

Source data

Extended Data Fig. 10 Integrated ACTA2 p.R179H and Marfan syndrome (MFS) aortic scRNAseq data.

A) Complete dataset following reciprocal PCA data integration demonstrating representation of all major cell clusters in both datasets. Red arrow indicates SMC cluster selected for downstream analysis. B) Expression plot of MYH11 confirming SMC identity for selected cluster. C) Overlaid UMAP plot for ACTA2 p.R179H and MFS SMCs demonstrating overrepresented sub-populations in ACTA2 patient (black arrows). D) Expression of ‘typical’ SMC modulation markers (FN1, COL1A1) with gradient of expression in both patients. E) Expression of representative altered SMC phenotypic trajectory markers in ACTA2 patient (see Fig. 7I) not activated in MFS SMCs. F) Activation of TGFβ1-responsive gene SERPINE1 is not observed in ACTA2 p.R179H SMCs as compared to MFS.

Supplementary information

Supplementary Information

Supplementary discussion, Tables 1 and 2, Figs. 1–7 and source data and western blots for supplementary figures.

Reporting Summary

Supplementary Data 1

Numerical source data for supplementary figures.

Supplementary Data 2

Image source data for supplementary figures.

Source data

Source Data Fig. 1

Unprocessed western blots, statistical source data and original image files.

Source Data Fig. 2

Unprocessed western blots, statistical source data and original image files.

Source Data Fig. 3

Unprocessed western blots and statistical source data.

Source Data Fig. 4

Unprocessed western blots, statistical source data and original image files.

Source Data Fig. 6

Unprocessed western blots, statistical source data and original image files.

Source Data Extended Data Fig./Table 1

Statistical source data.

Source Data Extended Data Fig./Table 2

Statistical source data and original image files.

Source Data Extended Data Fig./Table 3

Unprocessed western blots and statistical source data.

Source Data Extended Data Fig./Table 5

Statistical source data.

Source Data Extended Data Fig./Table 7

Unprocessed western blots and statistical source data.

Source Data Extended Data Fig./Table 8

Unprocessed western blots and statistical source data.

Source Data Extended Data Fig./Table 9

Unprocessed western blots and statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kwartler, C.S., Pedroza, A.J., Kaw, A. et al. Nuclear smooth muscle α-actin participates in vascular smooth muscle cell differentiation. Nat Cardiovasc Res 2, 937–955 (2023). https://doi.org/10.1038/s44161-023-00337-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s44161-023-00337-4

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