Abstract
Matrix-derived biophysical cues are known to regulate the activation of fibroblasts and their subsequent transdifferentiation into myofibroblasts1,2,3,4,5,6, but whether modulation of these signals can suppress fibrosis in intact tissues remains unclear, particularly in the cardiovascular system7,8,9,10. Here we demonstrate across multiple scales that inhibition of matrix mechanosensing in persistently activated cardiac fibroblasts potentiatesâin concert with soluble regulators of the TGFβ pathwayâa robust transcriptomic, morphological and metabolic shift towards quiescence. By conducting a meta-analysis of public human and mouse single-cell sequencing datasets, we identify the focal-adhesion-associated tyrosine kinase SRC as a fibroblast-enriched mechanosensor that can be targeted selectively in stromal cells to mimic the effects of matrix softening in vivo. Pharmacological inhibition of SRC by saracatinib, coupled with TGFβ suppression, induces synergistic repression of key profibrotic gene programs in fibroblasts, characterized by a marked inhibition of the MRTFâSRF pathway, which is not seen after treatment with either drug alone. Importantly, the dual treatment alleviates contractile dysfunction in fibrotic engineered heart tissues and in a mouse model of heart failure. Our findings point to joint inhibition of SRC-mediated stromal mechanosensing and TGFβ signalling as a potential mechanotherapeutic strategy for treating cardiovascular fibrosis.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 /Â 30Â days
cancel any time
Subscribe to this journal
Receive 51 print issues and online access
$199.00 per year
only $3.90 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout





Similar content being viewed by others
Data availability
Raw and processed sc/snRNA-seq data have been deposited in the NCBI Gene Expression Omnibus (GEO) under accession number GSE291370. Proteomics data generated from this study have been uploaded to ProteomeXchange and MassIVE under accession numbers PXD061188 and MSV000097212, respectively. Public datasets reanalysed in this study are cited in the references and are also available at NCBI GEO (GSE120064)39, the European Genomeâphenome Archive (EGA; EGAS00001006374)33, the Human Cell Atlas (HCA) Data Coordination Platform (DCP; ERP123138)36, the Broad Instituteâs Single Cell Portal (SCP498)37, the ArrayExpress database at EMBL-EBI (E-MTAB-7376 and E-MTAB-7365)38, ProteomeXchange (PXD016492)41, ProteomeXchange (PXD007171) and the NCBI National Library of Medicine (genome assemblies GRCh38, GCF_000001405.26 (human); and GRCm38, GCF_000001635.20 (mouse)). Source data are provided with this paper.
Code availability
No custom code or mathematical algorithms were developed for the study. All scRNA-seq/snRNA-seq data processing and analyses were performed using standard pipelines with Seurat (v.5.1.0) and open source R packages cited in the Methods and Reporting Summary, under the default settings provided in the corresponding vignettes.
References
Davis, J. & Molkentin, J. D. Myofibroblasts: trust your heart and let fate decide. J. Mol. Cell. Cardiol. 70, 9â18 (2014).
Herrera, J., Henke, C. A. & Bitterman, P. B. Extracellular matrix as a driver of progressive fibrosis. J. Clin. Invest. 128, 45â53 (2018).
Herum, K. M., Choppe, J., Kumar, A., Engler, A. J. & McCulloch, A. D. Mechanical regulation of cardiac fibroblast profibrotic phenotypes. Mol. Biol. Cell 28, 1871â1882 (2017).
Pesce, M. et al. Cardiac fibroblasts and mechanosensation in heart development, health and disease. Nat. Rev. Cardiol. 20, 309â324 (2023).
van Putten, S., Shafieyan, Y. & Hinz, B. Mechanical control of cardiac myofibroblasts. J. Mol. Cell. Cardiol. 93, 133â142 (2016).
Walker, C. J. et al. Nuclear mechanosensing drives chromatin remodelling in persistently activated fibroblasts. Nat. Biomed. Eng. 5, 1485â1499 (2021).
Frangogiannis, N. G. Can myocardial fibrosis be reversed? J. Am. Coll. Cardiol. 73, 2283â2285 (2019).
Nagaraju, C. K. et al. Myofibroblast phenotype and reversibility of fibrosis in patients with end-stage heart failure. J. Am. Coll. Cardiol. 73, 2267â2282 (2019).
Reichardt, I. M., Robeson, K. Z., Regnier, M. & Davis, J. Controlling cardiac fibrosis through fibroblast state space modulation. Cell Signal. 79, 109888 (2021).
Tschumperlin, D. J. & Lagares, D. Mechano-therapeutics: targeting mechanical signaling in fibrosis and tumor stroma. Pharmacol. Ther. 212, 107575 (2020).
Frangogiannis, N. G. Cardiac fibrosis. Cardiovasc. Res. 117, 1450â1488 (2021).
Travers, J. G., Kamal, F. A., Robbins, J., Yutzey, K. E. & Blaxall, B. C. Cardiac fibrosis: the fibroblast awakens. Circ. Res. 118, 1021â1040 (2016).
Sadek, H. & Olson, E. N. Toward the goal of human heart regeneration. Cell Stem Cell 26, 7â16 (2020).
Kong, P., Christia, P. & Frangogiannis, N. G. The pathogenesis of cardiac fibrosis. Cell. Mol. Life Sci. 71, 549â574 (2014).
Emig, R. et al. Passive myocardial mechanical properties: meaning, measurement, models. Biophys. Rev. 13, 587â610 (2021).
Yamamoto, K. et al. Myocardial stiffness is determined by ventricular fibrosis, but not by compensatory or excessive hypertrophy in hypertensive heart. Cardiovasc. Res. 55, 76â82 (2002).
Tallquist, M. D. & Molkentin, J. D. Redefining the identity of cardiac fibroblasts. Nat. Rev. Cardiol. 14, 484â491 (2017).
Ko, T. et al. Cardiac fibroblasts regulate the development of heart failure via Htra3-TGF-β-IGFBP7 axis. Nat. Commun. 13, 3275 (2022).
Wipff, P. J., Rifkin, D. B., Meister, J. J. & Hinz, B. Myofibroblast contraction activates latent TGF-β1 from the extracellular matrix. J. Cell Biol. 179, 1311â1323 (2007).
Francisco, J. et al. Blockade of fibroblast YAP attenuates cardiac fibrosis and dysfunction through MRTF-A inhibition. JACC Basic Transl. Sci. 5, 931â945 (2020).
Mia, M. M. et al. Loss of Yap/Taz in cardiac fibroblasts attenuates adverse remodelling and improves cardiac function. Cardiovasc. Res. 118, 1785â1804 (2022).
Revelo, X. S. et al. Cardiac resident macrophages prevent fibrosis and stimulate angiogenesis. Circ. Res. 129, 1086â1101 (2021).
Caliari, S. R. et al. Gradually softening hydrogels for modeling hepatic stellate cell behavior during fibrosis regression. Integr. Biol. 8, 720â728 (2016).
Driesen, R. B. et al. Reversible and irreversible differentiation of cardiac fibroblasts. Cardiovasc. Res. 101, 411â422 (2014).
Kollmannsberger, P., Bidan, C. M., Dunlop, J. W. C., Fratzl, P. & Vogel, V. Tensile forces drive a reversible fibroblast-to-myofibroblast transition during tissue growth in engineered clefts. Sci. Adv. 4, eaao4881 (2018).
Hall, C. et al. Chronic activation of human cardiac fibroblasts in vitro attenuates the reversibility of the myofibroblast phenotype. Sci. Rep. 13, 12137 (2023).
Li, C. X. et al. MicroRNA-21 preserves the fibrotic mechanical memory of mesenchymal stem cells. Nat. Mater. 16, 379â389 (2017).
Cho, S. et al. Mechanosensing by the lamina protects against nuclear rupture, DNA damage, and cell-cycle arrest. Dev. Cell 49, 920â935 e925 (2019).
Engler, A. J. et al. Embryonic cardiomyocytes beat best on a matrix with heart-like elasticity: scar-like rigidity inhibits beating. J. Cell Sci. 121, 3794â3802 (2008).
Gao, L. et al. Relationship between the efficacy of cardiac cell therapy and the inhibition of differentiation of human iPSC-derived nonmyocyte cardiac cells into myofibroblast-like cells. Circ. Res. 123, 1313â1325 (2018).
Cho, N., Razipour, S. E. & McCain, M. L. Featured Article: TGF-β1 dominates extracellular matrix rigidity for inducing differentiation of human cardiac fibroblasts to myofibroblasts. Exp. Biol. Med. 243, 601â612 (2018).
Speight, P., Kofler, M., Szaszi, K. & Kapus, A. Context-dependent switch in chemo/mechanotransduction via multilevel crosstalk among cytoskeleton-regulated MRTF and TAZ and TGFbeta-regulated Smad3. Nat. Commun. 7, 11642 (2016).
Reichart, D. et al. Pathogenic variants damage cell composition and single cell transcription in cardiomyopathies. Science 377, eabo1984 (2022).
Aldeiri, B. et al. Transgelin-expressing myofibroblasts orchestrate ventral midline closure through TGFβ signalling. Development 144, 3336â3348 (2017).
Madl, C. M., Flaig, I. A., Holbrook, C. A., Wang, Y. X. & Blau, H. M. Biophysical matrix cues from the regenerating niche direct muscle stem cell fate in engineered microenvironments. Biomaterials 275, 120973 (2021).
Litvinukova, M. et al. Cells of the adult human heart. Nature 588, 466â472 (2020).
Tucker, N. R. et al. Transcriptional and cellular diversity of the human heart. Circulation 142, 466â482 (2020).
Farbehi, N. et al. Single-cell expression profiling reveals dynamic flux of cardiac stromal, vascular and immune cells in health and injury. eLife 8, e43882 (2019).
Ren, Z. et al. Single-cell reconstruction of progression trajectory reveals intervention principles in pathological cardiac hypertrophy. Circulation 141, 1704â1719 (2020).
Totaro, A., Panciera, T. & Piccolo, S. YAP/TAZ upstream signals and downstream responses. Nat. Cell Biol. 20, 888â899 (2018).
Kuzmanov, U. et al. Mapping signalling perturbations in myocardial fibrosis via the integrative phosphoproteomic profiling of tissue from diverse sources. Nat. Biomed. Eng. 4, 889â900 (2020).
Subramanian, A. et al. A next generation connectivity map: L1000 platform and the first 1,000,000 profiles. Cell 171, 1437â1452 (2017).
Bugg, D. et al. MBNL1 drives dynamic transitions between fibroblasts and myofibroblasts in cardiac wound healing. Cell Stem Cell 29, 419â433 (2022).
Alexanian, M. et al. A transcriptional switch governs fibroblast activation in heart disease. Nature 595, 438â443 (2021).
Stowers, R. S. et al. Matrix stiffness induces a tumorigenic phenotype in mammary epithelium through changes in chromatin accessibility. Nat. Biomed. Eng. 3, 1009â1019 (2019).
Alexanian, M. et al. Chromatin remodelling drives immune cell-fibroblast communication in heart failure. Nature 635, 434â443 (2024).
Amrute, J. M. et al. Targeting immune-fibroblast cell communication in heart failure. Nature 635, 423â433 (2024).
Wang, J., Liu, S., Heallen, T. & Martin, J. F. The Hippo pathway in the heart: pivotal roles in development, disease, and regeneration. Nat. Rev. Cardiol. 15, 672â684 (2018).
Whitehead, A. J., Hocker, J. D., Ren, B. & Engler, A. J. Improved epicardial cardiac fibroblast generation from iPSCs. J. Mol. Cell. Cardiol. 164, 58â68 (2022).
Zhang, H., Shen, M. & Wu, J. C. Generation of quiescent cardiac fibroblasts derived from human induced pluripotent stem cells. Methods Mol. Biol. 2454, 109â115 (2022).
Burridge, P. W. et al. Chemically defined generation of human cardiomyocytes. Nat. Methods 11, 855â860 (2014).
Lian, X. et al. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc. Natl Acad. Sci. USA 109, E1848âE1857 (2012).
Street, K. et al. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genom. 19, 477 (2018).
La Manno, G. et al. RNA velocity of single cells. Nature 560, 494â498 (2018).
Alquicira-Hernandez, J. & Powell, J. E. Nebulosa recovers single-cell gene expression signals by kernel density estimation. Bioinformatics 37, 2485â2487 (2021).
Wu, T. et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation 2, 100141 (2021).
Jin, S. et al. Inference and analysis of cell-cell communication using CellChat. Nat. Commun. 12, 1088 (2021).
Holland, C. H. et al. Robustness and applicability of transcription factor and pathway analysis tools on single-cell RNA-seq data. Genome Biol. 21, 36 (2020).
Skowronek, P. et al. Rapid and in-depth coverage of the (phospho-)proteome with deep libraries and optimal window design for dia-PASEF. Mol. Cell. Proteom. 21, 100279 (2022).
Xu, W., Doshi, A., Lei, M., Eck, M. J. & Harrison, S. C. Crystal structures of c-Src reveal features of its autoinhibitory mechanism. Mol. Cell 3, 629â638 (1999).
Corsello, S. M. et al. The Drug Repurposing Hub: a next-generation drug library and information resource. Nat. Med. 23, 405â408 (2017).
Trott, O. & Olson, A. J. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem. 31, 455â461 (2010).
Daina, A., Michielin, O. & Zoete, V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci. Rep. 7, 42717 (2017).
Mannhardt, I. et al. Automated contraction analysis of human engineered heart tissue for cardiac drug safety screening. J. Vis. Exp. https://doi.org/10.3791/55461 (2017).
Chaturvedi, R. R. et al. Passive stiffness of myocardium from congenital heart disease and implications for diastole. Circulation 121, 979â988 (2010).
Rudebusch, J. et al. Dynamic adaptation of myocardial proteome during heart failure development. PLoS ONE 12, e0185915 (2017).
Acknowledgements
We thank the staff at the Vincent Coates Foundation Mass Spectrometry Laboratory, Stanford University Mass Spectrometry (SUMS, RRID: SCR_017801) and the Stanford Cancer Institute Proteomics/Mass Spectrometry Shared Resource for use of the Bruker timsTOF Ultra and the nanoElute 2 system (RRID: SCR_025639); and the staff at the Stanford Cardiovascular Institute for providing seed funding. This work was also supported by National Institutes of Health (NIH) grants F32 HL152483 and K99 HL166695 (to S.C.); F32 HL173968 (to A.C.); K99 HL163443 (to D.T.); and R01 HL113006, R01 HL130020, R01 HL141371, R01 HL141851, R01 HL150693, R01 HL145676, R01 HL146690 and R01 HL163680 (to J.C.W.), and by American Heart Association (AHA) 17MERIT33610009 (to J.C.W.).
Author information
Authors and Affiliations
Contributions
S.C. and J.C.W. conceptualized and designed the study. S.C., S.R. and J.C.W. wrote the manuscript. S.R. performed the animal experiments and helped to analyse data. C.M.M. performed the dynamic matrix softening experiments. A.C. performed the coIPâMS experiments and assisted in data analysis. H.K. and D.T. assisted with the EHT experiments. S.C. and A.K performed and analysed the FRAP experiments. A.M. performed the cardiomyocyte contractility assays. J.W.J. performed the Seahorse XF Assay experiments. H.S.S., D.T.P. and M.W. assisted with running, initial processing and integration of the scRNA-seq data. X.W. and P.N.T. performed the animal surgeries. M.M. performed the endothelial cell tube formation assays. N.M.B. assisted with RNA velocity analysis. J.L. and S.M. performed the virtual screen and MD simulations. V.D.W. provided the human foetal tissue samples. Y.J.W., H.M.B. and J.C.W. supervised the study and provided funding support.
Corresponding authors
Ethics declarations
Competing interests
J.C.W. is a co-founder and member of the scientific advisory board of Greenstone Biosciences. H.M.B. is a cofounder and member of the scientific advisory board of Epirium Bio. The other authors declare no competing interests.
Peer review
Peer review information
Nature thanks the anonymous reviewers for their contribution to the peer review of this work.
Additional information
Publisherâs note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data figures and tables
Extended Data Fig. 1 iPSC-CFs undergo spontaneous activation on stiff environments or at low cell densities unless mechanosensing is inhibited.
(A) Representative immunofluorescence images of αSMA (ACTA2) and YAP1 staining in iPSC-CFs cultured for ~10 passages on rigid tissue culture plastic (~âGPa) at varying cell densities (initial seeding at 2.5â40k cells/well). Images are representative of experiments repeated twice independently with similar results. Top row scale bar = 100âµm; bottom row scale bar = 10âµm. (B) % αSMA+ cells (left) and YAP1 N/C ratio (right) plotted versus cell seeding density: 2.5k, 5k, 10k, 20k, 40k cells/well: % αSMA+ cells (nâ=â13, 15, 15, 15, 20 randomly selected fields of view, respectively, from nâ=â3 biological replicates/group) and YAP1 N/C (nâ=â20, 18, 21, 26, 27 cells, respectively, from nâ=â3 biological replicates/group). (C) % αSMA+ cells plotted versus YAP1 N/C ratio (n-numbers equivalent to those of panel B). (D) αSMA immunoblot of iPSC-CFs undergoing spontaneous CF-to-MyoFb transition on hydrogels of varying stiffness for ~10 passages. Blots are representative of nâ=â2 independently performed experiments with similar results. (E) Reported stiffness ranges for embryonic, healthy adult, myocardial infarction (MI), heart failure with preserved ejection fraction (HFpEF), and transverse aortic constriction (TAC) hearts15,65. Normalized 2D projected cell area (top) and % of Ki67+ cells (bottom) versus stiffness: 2, 8, 16, 64âkPa and rigid plastic. Normalized cell area (nâ=â21, 18, 22, 13, 11 cells, respectively, from nâ=â3 biological replicates/group) and Ki67+ (nâ=â9, 15, 7, 5, 5, 8 randomly selected fields of view, respectively, from nâ=â3 biological replicates/group). (F) Half-maximal effective stiffness (âES50â, analogous to EC50 for a drug dose-response, in units of kPa) for each group, obtained from curve-fitting (data in main Fig. 1d, four-parameter dose-response stimulation with variable slope on GraphPad Prism v6.0). ES50 is defined as the gel stiffness at which the half-maximal response is produced, which in this case is the % of αSMA+ cells. ES50 for each group is plotted as individual bars, and the asymptotic standard error obtained from the best-fit curves (âStd. Errorâ values) are presented as error bars. P-values were calculated based on ordinary one-way ANOVA with Dunnettâs test (*âs) versus 2.5k/well (panel B), versus 2âkPa (panel E), or versus DMSO on plastic (panel E); and the two-sided Studentâs t-test (#âs). Data are meanâ±âSEM (with the exception of panel F, for which individual bars ± error bars represent ES50 ± Std. Error).
Extended Data Fig. 2 iPSC-CFs resemble primary human foetal CFs at the transcriptomic level.
(A) UMAP plot of quiescent iPSC-CFs mapped onto a reference dataset of primary fibroblasts (FBs) from various human foetal tissues (heart, lung, skin, kidney, and liver). (B) Feature plots showing expression of TCF21 and PDGFRA in iPSC-CFs and primary CFs. (C) Correlation plot of iPSC-CFs, FBs from various foetal tissues (lung, skin, kidney, liver), and major cardiac cell types (CFs, SMCs, CMs, and ECs). (D-F) Representative feature plots for: (D) pan-fibroblast genes, (E) CM marker genes, and (F) EC marker genes. (G) The 25 most variable genes along pseudotime inferred by Slingshot-based trajectory analysis of TGFβ-activated iPSC-CFs. Notable ECM genes (purple) and SMC/MyoFb-associated genes (orange) are highlighted. (H) Gene network plot (âcnetplotâ generated by clusterProfiler) for the MyoFb1 cluster.
Extended Data Fig. 3 Combination treatment drives morphological remodelling and population redistribution at the single-cell transcriptomic level without altering cell cycle.
(A) Additional representative time-lapse images of SM22α (TAGLN)-CFP reporter iPSC-derived MyoFbs subjected to light-induced matrix softening over 38 hrs, either with or without TGFβi. Scale bar = 100âµm. (B) Normalized 2D cell projected area and cell aspect ratio (length/width; L/W) before and 38 hrs after matrix softening. Stiff control (nâ=â10), TGFβi (nâ=â12), Softened (nâ=â11), Softened+ TGFβi (nâ=â10 cells). Data from the two separate experiments were normalized and pooled. (C) Individual UMAP plots (top row) and corresponding cell density plots (bottom row) for all four treatment groups. (D) UMAP plot generated based on the cell cycle phase of each cell, scored using canonical markers. (E) Cell cycle distribution of the total population in each of the four treatment conditions. (F) Cell cycle distribution by cluster in the four treatment conditions. (G) Representative immunofluorescence images of Ki67 and DAPI-stained MyoFbs. Bottom: quantitation of % Ki67+ cells in the four treatment conditions: Stiff control (nâ=â11), TGFβi (nâ=â5), Soft (nâ=â6), Soft+TGFβi (nâ=â5 randomly selected fields of view, from nâ=â3 biological replicates/group). (H) (i) RNA velocity plots of Stiff control (purple) and Soft+TGFβi (orange), and (ii) âÎRNA velocityâ generated by subtracting individual velocity vectors (\(\vec{{\rm{S}}{\rm{o}}{\rm{f}}{\rm{t}}+{\rm{T}}{\rm{G}}{\rm{F}}{\rm{\beta }}{\rm{i}}}-\vec{{\rm{S}}{\rm{t}}{\rm{i}}{\rm{f}}{\rm{f}}}\)) on the coordinate grid. Images in panels A and G are each representative of experiments repeated twice independently with similar results. P-values were calculated based on ordinary one-way ANOVA with Dunnettâs test (*âs) versus Stiff baseline control; and the two-sided Studentâs t-test (#âs). Data are meanâ±âSEM.
Extended Data Fig. 4 Combination treatment downregulates pro-fibrotic pathways and synergistically inhibits MRTF-A retention in the MyoFb nucleus.
(A) Heatmap of the top 25 TFs whose activity levels were most variable across the treatment conditions, for all identified clusters, based on activity fingerprinting by DoRothEA. (B) Violin plots of key downstream target genes of YAP/TAZ-TEAD. (C) Representative αSMA immunofluorescence images (left) and corresponding histogram of anti-αSMA intensity in single MyoFbs. Inset: bar graph of % αSMA+ cells (threshold set at 5-fold increase relative to baseline). Stiff (nâ=â49), TGFβi (nâ=â41), StiffâSoft (nâ=â22), StiffâSoft+ TGFβi (nâ=â30 randomly selected fields of view). Images are representative of experiments repeated nâ=â3 times independently with similar results. Scale bar = 100âµm. (D) Gel contraction assay of undifferentiated iPSC-CFs and MyoFbs after culturing (âprimingâ) for 4 days on soft or stiff substrates, with or without TGFβi treatment. Primed cells were trypsinized and mixed with a commercially available gel solution for the contraction assay. 2D gel areas were measured after 12âh (nâ=â3 biological replicates per group). (E) Dot plot showing the expression of major fibrosis-associated ECM proteins, ECM cross-linkers, and various MMP isoforms (collagenases and gelatinases) in the four groups after 2 days of treatment. (F) Schematic of experimental design illustrating TGFβ perturbations in a soft matrix background (orange), or stiff matrix culture in the presence of TGFβi. (G-H) Representative immunofluorescence images and quantitation of MRTF-A N/C ratio in the five treatment groups: Soft (nâ=â11), Stiff (nâ=â12), Stiff+LatB (latrunculin-B; nâ=â12), Soft+TGFβ (nâ=â11), Soft+TGFβâ+âSB (nâ=â11 cells). (I) Subcellular localization of enriched MRTF-A interactors in the CoIP-MS data. (J-K) Representative immunofluorescence images and quantitation of MRTF-A and SORBS2 co-localization in the nucleus and cytoplasm under the different treatment conditions: Undiff CF (nâ=â8), MyoFb (nâ=â37), TGFβi (nâ=â18), Soft (nâ=â12), Soft+TGFβi (nâ=â11 cells). Individual cells are shown as opaque data points in the background in panel K. Images in panels G and J are representative of experiments repeated twice independently with similar results. P-values were calculated based on ordinary one-way ANOVA with Dunnettâs test (*âs) versus Soft control (panel H); and the two-sided Studentâs t-test (#âs) versus MyoFb (panel K). Data are meanâ±âSEM.
Extended Data Fig. 5 SRC is unique among major cellular mechanosensors in its enrichment in cardiac stromal cells.
(A) Nebulosa feature plots for a broad range of major mechanosensors, including mechano-responsive TFs (e.g., YAP/TAZ), focal adhesion components (e.g., FAK (PTK2)), mechanically-gated ion channels (e.g., PIEZO2), integrins (e.g., ITGA1), and nuclear envelope proteins (e.g., LMNA) in adult mouse heart. SRCâs unique enrichment in CFs and mural cell populations is highlighted with arrowheads (magenta). (B-F) Representative UMAP plots and Nebulosa feature plots illustrating SRC expression in scRNA-seq datasets of: (B) human adult heart36, (C) FBs isolated from various human fetal tissues, (D) mouse model of myocardial infarction (MI)38, (E) additional mouse data set for TAC hearts39, and (F) TGFβ-simulated iPSC-CFs. (G) Violin plot of SRC expression across all clusters in TGFβ-simulated iPSC-CFs.
Extended Data Fig. 6 Saracatinib recapitulates the effects of matrix softening in vitro.
(A) Connectivity Map (CMap42) analysis showing the top 20 genes for which overexpression (OE) or knockdown (KD) results in similar perturbation signatures as saracatinib treatment. (B-C) Schematic of the experiment timeline (B) and representative immunofluorescence images (C) of cells treated with various drug combinations: âSARâ = saracatinib; âSBâ = SB431542; âPFDâ = pirfenidone; Scale bar = 150âµm; inset = 50âµm. (D) % αSMA-positive cells (defined as ⥠5X versus baseline control) over the course of the experiment. For days 2, 4, and 9: DMSO (nâ=â4, 11, 13), SAR (nâ=â2, 8, 10), SB (nâ=â3, 7, 8), PFD (nâ=â2, 8, 6), SARâ+âSB (nâ=â3, 6, 8), SARâ+âPFD (nâ=â3, 11, 14 randomly selected frames of view, respectively, from nâ=â3 biological replicates/group). (E-G) Cell aspect ratio (elongation) versus normalized 2D cell area (E), and nuclear/cytoplasmic ratio of (F) YAP1 and (G) MRTF-A at day 9. DMSO (nâ=â58), SAR (nâ=â44), SB (nâ=â41), PFD (nâ=â38), SARâ+âSB (nâ=â31), SARâ+âPFD (nâ=â44 cells). (H) Representative images of f-actin and DNA in cells treated with TGFβi, either alone or with three different SRC inhibitors (âDASâ = dasatinib; âBOSâ = bosutinib). Scale bar = 150âµm. (I) % cells with actomyosin stress fibres in the different treatment groups: CF, MyoFb, TGFβi, TGFβiâ+âDAS, TGFβiâ+âSAR, TGFβiâ+âBOS (nâ=â5, 8, 6, 5, 5, 5 randomly selected fields of view, respectively, from nâ=â3 biological replicates/group). (J) Scatter plot of cell aspect ratio (length/width; L/W) versus 2D area. CF (nâ=â35), MyoFb (nâ=â42), TGFβi (nâ=â69), TGFβiâ+âDAS (nâ=â25), TGFβiâ+âSAR (nâ=â35), TGFβiâ+âBOS (nâ=â77 cells). (K) Immunoblots and corresponding densitometry quantitation of pYAP1, αSMA, collagen-I, and POSTN. Blots are representative of nâ=â2 independently performed experiments with similar results (for collagen-I and POSTN, data from two additional experiments were normalized and pooled). Images shown are from two blots derived from the same samples from the same experiment, processed in parallel. For groups with nâ=â2, the error bars indicate the range between the two data points. (L) Subcellular localization of enriched MRTF-A interactors in the CoIP-MS data. (M) PCA analysis of CoIP-MS data from Fig. 3f, now including the SAR and SAR+TGFβi groups. (N) Heatmap of the anti-MRTF-A CoIP-MS data, showing all seven experimental groups (including those from Fig. 3fâh). The top 10 differentially enriched MRTF-A protein interactors in the MyoFb group (purple) are shown on the right. (O) Anti-SORBS2 immunoblot of the CoIP-MS input. The blot was not repeated due to sample availability after running the CoIP-MS. (P) Representative images (left) and quantitation (right) of MRTF-A and SORBS2 co-localization in the nucleus and cytoplasm across the different treatment groups (including those from Extended Data Fig. 4j,k. Undiff CF (nâ=â9), MyoFb (nâ=â37), TGFβi (nâ=â19), Soft (nâ=â12), Soft+TGFβi (nâ=â11), SAR (nâ=â11), SAR+TGFβi (nâ=â9 cells). Images in panels C, H, and P are representative of experiments repeated twice independently with similar results. P-values were calculated based on ordinary one-way ANOVA with Dunnettâs test (*âs) versus DMSO (panels D-G) or Undiff CF control (panel I); and the two-sided Studentâs t-test (#âs) versus MyoFb (panels J, P). Data are meanâ±âSEM.
Extended Data Fig. 7 Saracatinib potentiates reversal of metabolic states in MyoFbs without affecting the viability and function of cardiomyocytes and endothelial cells.
(A) Seahorse XF Assay measurements of: (i) oxygen consumption rate (OCR), (ii) extracellular acidification rate (ECAR), and (iii) OCR versus ECAR after 2 days of drug treatments (nâ=â4 replicates/condition). (B-i) PrestoBlue viability assay for varying concentrations of a direct YAP/TAZ inhibitor verteporfin (âVPâ, top) and the SRC inhibitor saracatinib (âSARâ, bottom) in iPSC-CMs, iPSC-ECs, iPSC-CFs, and iPSC-MyoFbs (nâ=â3 replicates per group). (B-ii) Half-maximal inhibitory concentration (IC50) values for VP and SAR, for each cell type. IC50 values were obtained from curve-fitting (data from panel B-ii, four-parameter dose-response inhibition with variable slope on GraphPad Prism v6.0). IC50 for each group is plotted as individual bars, and the asymptotic standard error obtained from the best-fit curves (âStd. Errorâ values) are presented as error bars. (C) Representative images (left) and quantitation (right) of EC tube formation assay after treatment with VP or SAR (1âµM each). nâ=â6 randomly selected fields of view, from nâ=â3 biological replicates/group. Images are representative of experiments repeated twice independently with similar results. (D) Heatmap (top) and line graph (bottom) illustrating the sharp decrease in SRC expression relative to cardiac genes (TTN, MYH7, and TNNT2) throughout the course of iPSC-CM differentiation (bulk RNA-seq, nâ=â3 technical replicates per time point). (E-G) Beating area, contraction velocity, and relaxation velocity measurements in VP- and SAR-treated iPSC-CMs at differentiation day 90 (D90): DMSO (nâ=â19), VP (nâ=â22), SAR (nâ=â24 randomly selected fields of view, from nâ=â3 biological replicates/group). (H) Western blot of phosphorylated SRC (Y416) and total SRC in MyoFbs under TGFβ drug perturbations. SAR was included as a positive control. Blots are representative of nâ=â3 experiments repeated with similar results. Images are from two blots from the same samples from the same experiment, processed in parallel. P-values were calculated based on ordinary one-way ANOVA with Dunnettâs test (*âs) versus DMSO control; and the two-sided Studentâs t-test (#âs). Data are meanâ±âSEM (with the exception of panel B-ii, for which individual bars ± error bars represent IC50 ± Std. Error).
Extended Data Fig. 8 Combination treatment alleviates contractile dysfunction in fibrotic engineered heart tissues.
(A) Schematic of EHT formation and fibrotic compaction of EHTs upon TGFβ stimulation. Right inset: snapshots of relaxed (diastole; cyan) and contracted (systole; magenta) states. (B) Representative images of EHTs for each condition in their relaxed states (diastole). The distance between the centre of the two silicon posts were used to measure the diastolic tissue length, Ld (yellow dotted line). Images are representative of experiments repeated four times with similar results. Scale bar = 1âmm. (C-F) Fractional shortening (FS%) and diastolic tissue length (Ld) measured over time for each drug treatment group (C,E) and FS% and Ld after 2 days of treatment with the different drugs, normalized to DMSO control (D,F). Ctrl (nâ=â13), TGFβ (nâ=â13), S (nâ=â10), P (nâ=â11), Sâ+âP (nâ=â12 EHTs). (G) Elastic modulus (stiffness) of EHTs 4 days after the drug treatments: Ctrl (nâ=â5), TGFβ (nâ=â7), S (nâ=â4), P (nâ=â3), Sâ+âP (nâ=â7). For panels C and E (between days 0 and 6) and for panels D and G, data from four separate experiments were normalized and pooled. P-values were calculated based on ordinary one-way ANOVA with Dunnettâs test (*âs) versus control EHTs; and the two-sided Studentâs t-test (#âs). Data are meanâ±âSEM.
Extended Data Fig. 9 Combination treatment in MI and TAC models of cardiac fibrosis.
(A) Schematic summary of the MI experiment. SARâ+âPFD (âSâ+âPâ) dual treatment was started at three different time points: Sham (nâ=â6), MI (nâ=â14), and Sâ+âP starting on day 1 (nâ=â8), day 4 (nâ=â10), and day 7 (nâ=â4 animals) post-MI surgery. (B) Survival curves for the five treatment groups. (C) Representative images of Massonâs trichrome-stained tissue sections and (D) M-mode echocardiography scans for each group at day 21 post-MI. Images are representative of at least four animals per group, from one MI cohort. (E-I) Time-course of left ventricular ejection fraction, LVEF (%, (E)), and endpoint echocardiography measurements of LVEF (%) (F), fractional shortening (FS%, (G)), left ventricular posterior wall thickness in diastole (LPWd, (H)), and left ventricular internal diameter (LVIDd, (I)). At endpoint, Sham (nâ=â6), MI (nâ=â9), âSâ+âP (d4-)â (nâ=â8), and âSâ+âP (d7-)â (nâ=â4 animals). (J) Schematic summary of the TAC experiment, copied from Fig. 5b. (K-M) Time-course measurements of FS (K, left; for days 18, 24, and 42, data from three separate experiments were normalized and pooled), and week 9 measurements of: FS (K, right), left ventricular internal diameter (LVIDd, (L)), and heart weight/body weight (mg/g) ratio (M). Sham, TAC, Sâ+âP, Sâ+âP withdrawn, S, and P (nâ=â10, 12, 12, 6, 7, 7 animals, respectively). P-values were calculated based on ordinary one-way ANOVA with Dunnettâs test (*âs) versus Sham; and the two-sided Studentâs t-test (#âs) versus MI (panels F-I) or versus TAC (panels K-M). Data are meanâ±âSEM.
Extended Data Fig. 10 Combination treatment induces transcriptional and protein-level changes in TAC hearts without significantly affecting inflammation or immune signalling.
(A) Left: Western blot of phosphorylated YAP (S127) and total YAP in whole heart lysates from the different drug treatment groups: Sham, Sham Sâ+âP, TAC 4 wk, TAC 6 wk, Sâ+âP, S, P (nâ=â5, 5, 10, 9, 9, 8, 9 hearts per lysate, respectively). Right: snRNA-seq violin plot displaying module scores for major YAP/TAZ target genes (Ccn2 (CTGF), Ankrd1, Amotl2, Birc5, and Igfbp2) specifically in CFs. (B) Violin plots showing module scores for common genes associated with oxygen consumption rate (OCR, left) and extracellular acidification rate (ECAR, right). (C) Normalized mass spectrometry intensity values for SORBS2 (ArgBP2) in two published datasets of mouse TAC hearts41,66: Kuzmanov et al and Rudebusch et al. (nâ=â5 and 4 tissue samples, respectively). (D) Violin plot of Sorbs2 expression in the five groups. Horizontal bars indicate mean. Cells with >0 Sorbs2 expression are shown. (E) Gene Ontology enrichment analysis for SARâ+âPFD versus TAC hearts. Fisherâs exact test (with Benjamini-Hochberg adjustment; one-sided). (F) Dot plot of major pro-inflammatory cytokine genes in the heart. (G-I) CellChat-based inference of cell-cell communication mechanisms, showing changes in modes of signalling from immune cells to CF populations (immuneâCF) in the SARâ+âPFD group relative to TAC (H,I). A permutation test (randomization; one-sided) was used to compute communication probability strength of ligand-receptor pairs. (J-K) Immunoblots and corresponding quantitation of SRC phosphorylation and key fibrosis-associated proteins αSMA and POSTN for each group: Sham, Sham Sâ+âP, TAC (4 wk), TAC (6 wk), Sâ+âP, S, P (nâ=â5, 5, 10, 9, 9, 8, 9 hearts per lysate, respectively). Immunoblots in panels A and K are representative of nâ=â3 experiments performed independently with similar results. Images are from two blots from the same samples from the same experiment, processed under identical conditions. For panel K, data from an additional experiment was normalized and pooled. P-values were calculated based on ordinary one-way ANOVA with Dunnettâs test (*âs) versus Sham; and the two-sided Studentâs t-test (#âs). Data are meanâ±âSEM.
Supplementary information
Supplementary Fig. 1
Immunoblot source data. Original uncropped immunoblot images for the main figures (pages 1â2) and extended data figures (pages 3â4). The red dotted boxes and text indicate the exact cropped regions that were included in the figures and the corresponding probed protein, respectively. The black text labels to the right of each blot indicate proteins that were probed simultaneously (for example, for GAPDH normalization) but not included in the figures. Uncropped images for: Fig. 3e (4 samples, from left to right: soft, softâ+âTGFβi, stiff (Ctrl), stiffâ+âTGFβi), three separate immunoblots of the same samples run under identical conditions are shown (a); Fig. 4f (3 samples, from left to right: sham, TAC (4 weeks), TAC (6 weeks)) (b); Extended Data Fig. 1d (4 samples, from left to right: 2, 8, 16 and 64 kPa) (c); Extended Data Fig. 6k (6 samples, from left to right: Undiff CF, MyoFb, SARâ+âSB, SARâ+âPFD, SB, PFD), two separate immunoblots of the same samples run in parallel are shown (d); Extended Data Fig. 6o (7 samples, from left to right: Undiff CF, MyoFb, SAR, TGFβi, SARâ+âTGFβi, soft, softâ+âTGFβi) (e); Extended Data Fig. 7h (4 samples, from left to right: DMSO, SB (TGFβi), PFD (TGFβi), SAR; the bottom blot is identical to the one shown on page 1 (second row, top left) with two shared samples from the same experimental conditions) (f); and Extended Data Fig. 10a and 10j, respectively (6 samples, from left to right: sham, sham Sâ+âP, TAC (4 weeks), TAC (6 weeks), Sâ+âP, S, P) (g and h). The rightward arrows (labelled with strip and re-blot) indicate that the blot (left) was first stripped then reprobed using a different antibody resulting in a new blot (right) of the same samples from the same experiment.
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.
About this article
Cite this article
Cho, S., Rhee, S., Madl, C.M. et al. Selective inhibition of stromal mechanosensing suppresses cardiac fibrosis. Nature 642, 766â775 (2025). https://doi.org/10.1038/s41586-025-08945-9
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41586-025-08945-9
This article is cited by
-
FAERS based disproportionality analysis and network pharmacology investigation of doxorubicin associated drug induced cardiotoxicity
Scientific Reports (2025)
-
Dual mechanotherapy for the treatment of cardiac fibrosis
Nature Reviews Cardiology (2025)
-
Towards Therapeutic Targeting of Fibroblasts in Giant Cell Arteritis
Current Treatment Options in Rheumatology (2025)
-
Durotaxis is a driver and potential therapeutic target in lung fibrosis and metastatic pancreatic cancer
Nature Cell Biology (2025)


