1
|
Ramli MFH, Aguado BA, Young JL. Signals from the extracellular matrix: Region- and sex-specificity in cardiac aging. Curr Opin Cell Biol 2025; 95:102524. [PMID: 40347709 DOI: 10.1016/j.ceb.2025.102524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 05/14/2025]
Abstract
During aging, the cardiac extracellular matrix (ECM) undergoes gradual remodeling that reduces the heart's ability to function. Specific ECM changes cause alterations in cellular signaling pathways, eliciting maladaptive responses. Here, we provide insight into the current knowledge of how age-specific ECM changes contribute to altered ligand-receptor interactions, dysregulated mechanotransduction, and the propagation of pro-fibrotic signaling cascades that underpin dysfunction. We also highlight regional and sex differences that new biomolecular and bioengineered technologies have recently uncovered. We call for new biomaterial strategies that mimic spatiotemporal and sex-specific ECM alterations to equip researchers with the tools to unravel complex cellular signaling events. We believe this can be achieved through interdisciplinary cooperation amongst researchers spanning matrix biology, biomaterials, spatial omics, and biomedical engineering.
Collapse
Affiliation(s)
- Md Faris H Ramli
- Mechanobiology Institute (MBI), National University of Singapore, 117411, Singapore
| | - Brian A Aguado
- Shu Chien-Gene Lay Department of Bioengineering, University of California, La Jolla, San Diego, CA, 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA; Program in Materials Science and Engineering, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Jennifer L Young
- Mechanobiology Institute (MBI), National University of Singapore, 117411, Singapore; Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 117583, Singapore.
| |
Collapse
|
2
|
Frangogiannis NG. Targeting macrophage-fibroblast interactions in the failing heart. Nat Rev Cardiol 2025; 22:223-224. [PMID: 39681733 DOI: 10.1038/s41569-024-01112-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute. Albert Einstein College of Medicine, Bronx, NY, USA.
- Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA.
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
3
|
Li G, Ni C, Wang J, Zhang F, Fu Z, Wang L, Wang B, Liu Y, Zhao J, Li M, Lin H, Liao F, Ye S, Zhang Y, Cai J, Shi S, Zhong Z, Shi Y, He J, Xiong X, Xu Y, Chen J, Zhu W, Wang Y, Wang J, Hu X. Dynamic molecular atlas of cardiac fibrosis at single-cell resolution shows CD248 in cardiac fibroblasts orchestrates interactions with immune cells. NATURE CARDIOVASCULAR RESEARCH 2025; 4:380-396. [PMID: 40148545 DOI: 10.1038/s44161-025-00617-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 01/30/2025] [Indexed: 03/29/2025]
Abstract
Post-injury remodeling is a complex process involving temporal specific cellular interactions in the injured tissue where the resident fibroblasts play multiple roles. Here, we performed single-cell and spatial transcriptome analysis in human and mouse infarcted hearts to dissect the molecular basis of these interactions. We identified a unique fibroblast subset with high CD248 expression, strongly associated with extracellular matrix remodeling. Genetic Cd248 deletion in fibroblasts mitigated cardiac fibrosis and dysfunction following ischemia/reperfusion. Mechanistically, CD248 stabilizes type I transforming growth factor beta receptor and thus upregulates fibroblast ACKR3 expression, leading to enhanced T cell retention. This CD248-mediated fibroblast-T cell interaction is required to sustain fibroblast activation and scar expansion. Disrupting this interaction using monoclonal antibody or chimeric antigen receptor T cell reduces T cell infiltration and consequently ameliorates cardiac fibrosis and dysfunction. Our findings reveal a CD248+ fibroblast subpopulation as a key regulator of immune-fibroblast cross-talk and a potential therapy to treat tissue fibrosis.
Collapse
Affiliation(s)
- Guohua Li
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Cheng Ni
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Jiacheng Wang
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Feimu Zhang
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Zaiyang Fu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Lingjun Wang
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Biqing Wang
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Ye Liu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Jing Zhao
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Mo Li
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Hao Lin
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Fei Liao
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Shuchang Ye
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Yu Zhang
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Jiayue Cai
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Shaohui Shi
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Zhiwei Zhong
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Yanna Shi
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Junhua He
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Xushen Xiong
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Yang Xu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Jinghai Chen
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Wei Zhu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Yibin Wang
- Programme in Cardiovascular and Metabolic Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Jian'an Wang
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, China
| | - Xinyang Hu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China.
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, China.
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, China.
| |
Collapse
|
4
|
Zhang J, Wang W, Wang Z, Zhou M, Wu S. Deciphering myocardial fibrosis: a comprehensive bibliometric analysis of mechanism over the period 1992-2023. J Cardiothorac Surg 2025; 20:170. [PMID: 40158166 PMCID: PMC11954359 DOI: 10.1186/s13019-025-03404-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 03/13/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Myocardial fibrosis is a critical link in preventing the progression of heart disease. This study conducted a bibliometric analysis of its mechanism to identify trends and hotspots, aiming to provide valuable references for heart disease prevention and treatment. METHODS This research relies on the Web of Science Core Collection, capturing all related publications on the mechanism of myocardial fibrosis up to November 11, 2023. For the bibliometric analysis, CiteSpace 6.2.R5 (64-bit) and VOSviewer 1.6.19 software tools were utilized. RESULTS The mechanism of myocardial fibrosis research involves 14,931 authors from 2,370 institutions in 71 countries/regions, resulting in 2,431 published studies. Nattel Stanley is the most prolific author, while Francogianis Ng is noted for the highest co-publication frequency. The United States leads in countries/regions, with the University of California System being the top institution. Cardiovascular Research is a primary outlet for new studies, and Circulation is a key reference in this research community. Current research primarily examines how myocardial fibrosis contributes to heart failure, myocardial infarction, and myocardial hypertrophy. This emerging field also explores the role of fibroblasts in myocardial injury and investigates innovative treatments to reduce myocardial fibrosis. CONCLUSIONS Preventing myocardial fibrosis is a crucial strategy in the fight against heart disease. This study utilises bibliometric analysis to explore the vast array of literature on the mechanism of myocardial fibrosis, mapping the research landscape and provide literature references for potential breakthroughs in heart disease prevention and treatment strategies.
Collapse
Affiliation(s)
- Jiaojiao Zhang
- School of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, Anhui Province, 230000, China
| | - Wenhui Wang
- School of Acupuncture, Moxibustion and Tuina, Anhui University of Chinese Medicine, Hefei, Anhui Province, 230000, China
| | - Zhen Wang
- Bozhou Huatuo Hospital of Traditional Chinese Medicine, Bozhou, Anhui Province, 236800, China
| | - Meiqi Zhou
- School of Acupuncture, Moxibustion and Tuina, Anhui University of Chinese Medicine, Hefei, Anhui Province, 230000, China.
- Acupuncture and Meridian Research Institute, Anhui Academy of Chinese Medicine, Hefei, Anhui Province, 230000, China.
- Key Laboratory of Meridian Viscera Correlationship, Anhui Academy of Chinese Medicine, Hefei, Anhui Province, 230000, China.
| | - Shengbing Wu
- School of Acupuncture, Moxibustion and Tuina, Anhui University of Chinese Medicine, Hefei, Anhui Province, 230000, China.
- Acupuncture and Meridian Research Institute, Anhui Academy of Chinese Medicine, Hefei, Anhui Province, 230000, China.
- Key Laboratory of Meridian Viscera Correlationship, Anhui Academy of Chinese Medicine, Hefei, Anhui Province, 230000, China.
| |
Collapse
|
5
|
Ghazal R, Wang M, Liu D, Tschumperlin DJ, Pereira NL. Cardiac Fibrosis in the Multi-Omics Era: Implications for Heart Failure. Circ Res 2025; 136:773-802. [PMID: 40146800 PMCID: PMC11949229 DOI: 10.1161/circresaha.124.325402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Cardiac fibrosis, a hallmark of heart failure and various cardiomyopathies, represents a complex pathological process that has long challenged therapeutic intervention. High-throughput omics technologies have begun revolutionizing our understanding of the molecular mechanisms driving cardiac fibrosis and are providing unprecedented insights into its heterogeneity and progression. This review provides a comprehensive analysis of how techniques-encompassing genomics, epigenomics, transcriptomics, proteomics, and metabolomics-are providing insight into our understanding of cardiac fibrosis. Genomic studies have identified novel genetic variants and regulatory networks associated with fibrosis susceptibility and progression, and single-cell transcriptomics has unveiled distinct cardiac fibroblast subpopulations with unique molecular signatures. Epigenomic profiling has revealed dynamic chromatin modifications controlling fibroblast activation states, and proteomic analyses have identified novel biomarkers and potential therapeutic targets. Metabolomic studies have uncovered important alterations in cardiac energetics and substrate utilization during fibrotic remodeling. The integration of these multi-omic data sets has led to the identification of previously unrecognized pathogenic mechanisms and potential therapeutic targets, including cell-type-specific interventions and metabolic modulators. We discuss how these advances are driving the development of precision medicine approaches for cardiac fibrosis while highlighting current challenges and future directions in translating multi-omic insights into effective therapeutic strategies. This review provides a systems-level perspective on cardiac fibrosis that may inform the development of more effective, personalized therapeutic approaches for heart failure and related cardiovascular diseases.
Collapse
Affiliation(s)
- Rachad Ghazal
- Departments of Cardiovascular Diseases (R.G., N.L.P.), Mayo Clinic, Rochester, MN
| | - Min Wang
- Molecular Pharmacology and Experimental Therapeutics (M.W., D.L., N.L.P.), Mayo Clinic, Rochester, MN
| | - Duan Liu
- Molecular Pharmacology and Experimental Therapeutics (M.W., D.L., N.L.P.), Mayo Clinic, Rochester, MN
| | | | - Naveen L. Pereira
- Departments of Cardiovascular Diseases (R.G., N.L.P.), Mayo Clinic, Rochester, MN
- Molecular Pharmacology and Experimental Therapeutics (M.W., D.L., N.L.P.), Mayo Clinic, Rochester, MN
| |
Collapse
|
6
|
Duan X, Zhang L, Liu K, Guo K, You Y, Jia H, Zhou S, Han B. Macrophage-derived SPP1 exacerbate myocardial injury by interacting with fibroblasts in viral myocarditis. Biol Direct 2025; 20:30. [PMID: 40087693 PMCID: PMC11907792 DOI: 10.1186/s13062-025-00621-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 02/22/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Viral myocarditis (VMC) is an inflammatory myocardial condition triggered by viral infections which involves pathogenic-related damage and immune-mediated damage. However, the precise immunopathogenic mechanisms underlying VMC remain elusive. METHODS We performed single-cell RNA sequencing on mouse hearts during the acute phase of CVB3-induced VMC. After manually annotating cell types, functional analyses of macrophage were performed by cell ratio changes, customized gene set module scoring and CellPhoneDB. Utilizing indirect co-culture experiments in vitro, the effects of macrophage-derived SPP1 on cardiac fibroblasts were investigated. Depletion of macrophages and inhibition of SPP1 expression in mice were carried out to study the effects of macrophage-derived SPP1 on cardiac function, inflammation levels, and myocardial injury in mice with VMC. RESULTS Our data revealed that macrophages are the major immune cells which infiltrate the heart during the acute phase of VMC, particularly a macrophage subpopulation which highly expresses Spp1 (Spp1+ macrophages) and exhibited characteristics of peripheral blood monocytes. Spp1+ macrophages communicate extensively with fibroblasts during VMC, and that SPP1 promotes fibroblast conversion to an inflammatory phenotype with high Ccl2/Ccl7 expression. This in turn increases monocyte chemotaxis to the heart. Besides, a partial depletion of macrophages in the early stages of VMC attenuated myocardial inflammation and myocardial injury in mice. Inhibition of SPP1 reduced cardiac macrophage infiltration, attenuated myocardial inflammation, and improved cardiac function in VMC mice. CONCLUSION Our findings suggested that Spp1+ macrophages could self-recruit, and macrophage-derived SPP1 exacerbated myocardial immune injury by promoting high Ccl2/Ccl7 expression in fibroblasts. Our study advances understandings of VMC pathogenesis, and provides novel insight into potential immunotherapies for VMC.
Collapse
Affiliation(s)
- Xiuyun Duan
- Department of Pediatric Cardiology, Cheeloo Colledge of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Li Zhang
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Children's Health and Disease office, Shandong Provincial Hospital, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Keyu Liu
- Department of Pediatric Cardiology, Cheeloo Colledge of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Kaiyin Guo
- Rui Jin Hospital Nanxiang Branch, Shanghai, 201802, China
| | - Yingnan You
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Children's Health and Disease office, Shandong Provincial Hospital, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Hailin Jia
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Children's Health and Disease office, Shandong Provincial Hospital, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shan Zhou
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Children's Health and Disease office, Shandong Provincial Hospital, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Bo Han
- Department of Pediatric Cardiology, Cheeloo Colledge of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong Provincial Clinical Research Center for Children's Health and Disease office, Shandong Provincial Hospital, Jinan, Shandong, China.
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
7
|
Amancherla K, Taravella Oill AM, Bledsoe X, Williams AL, Chow N, Zhao S, Sheng Q, Bearl DW, Hoffman RD, Menachem JN, Siddiqi HK, Brinkley DM, Mee ED, Hadad N, Agrawal V, Schmeckpepper J, Rali AS, Tsai S, Farber-Eger EH, Wells QS, Freedman JE, Tucker NR, Schlendorf KH, Gamazon ER, Shah RV, Banovich N. Dynamic responses to rejection in the transplanted human heart revealed through spatial transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640852. [PMID: 40093136 PMCID: PMC11908199 DOI: 10.1101/2025.02.28.640852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Allograft rejection following solid-organ transplantation is a major cause of graft dysfunction and mortality. Current approaches to diagnosis rely on histology, which exhibits wide diagnostic variability and lacks access to molecular phenotypes that may stratify therapeutic response. Here, we leverage image-based spatial transcriptomics at sub-cellular resolution in longitudinal human cardiac biopsies to characterize transcriptional heterogeneity in 62 adult and pediatric heart transplant (HT) recipients during and following histologically-diagnosed rejection. Across 28 cell types, we identified significant differences in abundance in CD4 + and CD8 + T cells, fibroblasts, and endothelial cells across different biological classes of rejection (cellular, mixed, antibody-mediated). We observed a broad overlap in cellular transcriptional states across histologic rejection severity and biological class and significant heterogeneity within rejection severity grades that would qualify for immunomodulatory treatment. Individuals who had resolved rejection after therapy had a distinct transcriptomic profile relative to those with persistent rejection, including 216 genes across 6 cell types along pathways of inflammation, IL6-JAK-STAT3 signaling, IFNα/IFNγ response, and TNFα signaling. Spatial transcriptomics also identified genes linked to long-term prognostic outcomes post-HT. These results underscore importance of subtyping immunologic states during rejection to stratify immune-cardiac interactions following HT that are therapeutically relevant to short- and long-term rejection-related outcomes.
Collapse
|
8
|
Kel A, Thum T, Kunduzova O. Targeting fibroblast phenotype switching in cardiac remodelling as a promising antifibrotic strategy. Eur Heart J 2025; 46:354-358. [PMID: 39582108 DOI: 10.1093/eurheartj/ehae722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/16/2024] [Accepted: 10/06/2024] [Indexed: 11/26/2024] Open
Abstract
Myocardial fibrosis, a common feature of heart disease, remains an unsolved clinical challenge. Fibrosis resolution requires activation of cardiac fibroblasts exhibiting context-dependent beneficial and detrimental dichotomy. Here, we explored the hypothesis of fibroblast reversible transition between quiescence and activated myofibroblastic states as a manifestation of cell phenotypic switching in myocardial remodelling. In support, gene regulatory networks executing conversion of cardiac fibroblasts to myofibroblasts and vice versa in fibrosis resolution are reconstructed using TRANSPATH database. In a scenario of fibroblast activation triggered by transforming growth factor β, a cardinal mediator of tissue fibrosis, signalling cascades governing entry into or exit from specific fibroblast statures in cardiac fibrotic remodelling were dissected. It is suggested that fibroblast phenotypic switching constitutes the central gait toward guiding cell state-gating strategies to counteract adverse cardiac fibrosis, a devastating disorder with no approved therapeutic option.
Collapse
Affiliation(s)
- Alexander Kel
- R&D Department, GeneXplain GmbH, Wolfenbüttel 38302, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
- Center for Translational Regenerative Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Oksana Kunduzova
- Institute of Metabolic and Cardiovascular Diseases (I2MC), National Institute of Health and Medical Research (INSERM) 1297, Toulouse III University, 1 Avenue J. Poulhes, Toulouse 31432, France
| |
Collapse
|
9
|
Li N, Zhu C, Xue Y, Chen N, Xu W, Song M, Qi M, Huang S, Fang M. The matrix protease ADAMTS1 is transcriptionally activated by KLF6 and contributes to cardiac fibrosis in non-ischemic cardiomyopathy. Life Sci 2025; 361:123295. [PMID: 39643035 DOI: 10.1016/j.lfs.2024.123295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/16/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
AIMS Aberrant cardiac fibrosis, defined as excessive production and deposition of extracellular matrix (ECM), is mediated by myofibroblasts. ECM-producing myofibroblasts are primarily derived from resident fibroblasts during cardiac fibrosis. The mechanism underlying fibroblast-myofibroblast transition is not fully understood. METHODS Cardiac fibrosis was induced by transverse aortic constriction (TAC) or by angiotensin II (Ang II) infusion in C57B6/j mice. Cellular transcriptome was evaluated by RNA-seq and CUT&Tag-seq. RESULTS Integrated transcriptomic screening revealed that a disintegrin and metalloproteinase with thrombospondin motifs 1 (ADAMTS1) was a novel transcriptional target for Kruppel-like factor 6 (KLF6) in cardiac fibroblasts. Treatment with either TGF-β or Ang II up-regulated ADAMTS1 expression. KLF6 knockdown attenuated whereas KLF6 over-expression enhanced ADAMTS1 induction. ChIP assay and reporter assay showed that KLF6 was recruited to the ADAMTS1 promoter to activate its transcription. Consistently, ADAMTS1 knockdown suppressed fibroblast-myofibroblast transition in vitro. Importantly, myofibroblast-specific ADAMTS1 depletion attenuated cardiac fibrosis and normalized heart function in mice. SIGNIFICANCE In conclusion, our data demonstrate that ADAMTS1, as a downstream target of KLF6, contributes to cardiac fibrosis by regulating fibroblast-myofibroblast transition.
Collapse
Affiliation(s)
- Nan Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Human Anatomy, Nanjing Medical University, Nanjing, China
| | - Chenghao Zhu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Yujia Xue
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Naxia Chen
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research and Key Laboratory of Emergency and Trauma of Ministry of Education, Institute of Cardiovascular Research, Department of Cardiology, the First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Wenping Xu
- Center for Experimental Medicine, Jiangsu Health Vocational College, Nanjing, China
| | - Mingzi Song
- Center for Experimental Medicine, Jiangsu Health Vocational College, Nanjing, China
| | - Mengwen Qi
- Center for Experimental Medicine, Jiangsu Health Vocational College, Nanjing, China
| | - Shan Huang
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research and Key Laboratory of Emergency and Trauma of Ministry of Education, Institute of Cardiovascular Research, Department of Cardiology, the First Affiliated Hospital, Hainan Medical University, Haikou, China.
| | - Mingming Fang
- Center for Experimental Medicine, Jiangsu Health Vocational College, Nanjing, China.
| |
Collapse
|
10
|
Song Y, Wang L, Wang H, Ma H, Xu J, Liu J, Qian L. Decoding aging in the heart via single cell dual omics of non-cardiomyocytes. iScience 2024; 27:111469. [PMID: 39735437 PMCID: PMC11681900 DOI: 10.1016/j.isci.2024.111469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/30/2024] [Accepted: 11/21/2024] [Indexed: 12/31/2024] Open
Abstract
To understand heart aging at the single-cell level, we employed single-cell dual omics (scRNA-seq and scATAC-seq) in profiling non-myocytes (non-CMs) from young, middle-aged, and elderly mice. Non-CMs, vital in heart development, physiology, and pathology, are understudied compared to cardiomyocytes. Our analysis revealed aging response heterogeneity and its dynamics over time. Immune cells, notably macrophages and neutrophils, showed significant aging alterations, while endothelial cells displayed moderate changes. We identified distinct aging signatures within the cell type, including differential gene expression, transcription factor activity, and motif variation. Sub-cluster analysis revealed intra-cell type heterogeneity, characterized by diverse aging patterns. The senescence-associated secretory phenotype emerged as a key aging-related phenotype. Moreover, aging significantly influenced cell-cell communication, especially impacting a fibroblast sub-cluster with high expression of ERBB4. This study elucidates the complex cellular and molecular landscape of cardiac aging and offers guidance for potential therapeutic avenues to treat aging-related heart diseases.
Collapse
Affiliation(s)
- Yiran Song
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Li Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Haofei Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hong Ma
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jun Xu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
11
|
Rudman-Melnick V, Vanhoutte D, Stowers K, Sargent M, Adam M, Ma Q, Perl AKT, Miethke AG, Burg A, Shi T, Hildeman DA, Woodle ESS, Kofron JM, Devarajan P. Gucy1α1 specifically marks kidney, heart, lung and liver fibroblasts. Sci Rep 2024; 14:29307. [PMID: 39592775 PMCID: PMC11599588 DOI: 10.1038/s41598-024-80930-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/22/2024] [Indexed: 11/28/2024] Open
Abstract
Fibrosis is a common outcome of numerous pathologies, including chronic kidney disease (CKD), a progressive renal function deterioration. Current approaches to target activated fibroblasts, key effector contributors to fibrotic tissue remodeling, lack specificity. Here, we report Gucy1α1 as a specific kidney fibroblast marker. Gucy1α1 levels significantly increased over the course of two clinically relevant murine CKD models and directly correlated with established fibrosis markers. Immunofluorescent (IF) imaging showed that Gucy1α1 comprehensively labelled cortical and medullary quiescent and activated fibroblasts in the control kidney and throughout injury progression, respectively. Unlike traditionally used markers platelet derived growth factor receptor beta (Pdgfrβ) and vimentin (Vim), Gucy1α1 did not overlap with off-target populations such as podocytes. Notably, Gucy1α1 labelled kidney fibroblasts in both male and female mice. Furthermore, we observed elevated GUCY1α1 expression in the human fibrotic kidney and lung. Studies in the murine models of cardiac and liver fibrosis revealed Gucy1α1 elevation in activated Pdgfrβ-, Vim- and alpha smooth muscle actin (αSma)-expressing fibroblasts paralleling injury progression and resolution. Overall, we demonstrate Gucy1α1 as an exclusive fibroblast marker in both sexes. Due to its multiorgan translational potential, GUCY1α1 might provide a novel promising strategy to specifically target and mechanistically examine fibroblasts.
Collapse
Affiliation(s)
- Valeria Rudman-Melnick
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, 268-280 Albert Sabin Way, location T, floor 6, suite 272, Cincinnati, OH, 45229, USA
| | - Davy Vanhoutte
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kaitlynn Stowers
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, 268-280 Albert Sabin Way, location T, floor 6, suite 272, Cincinnati, OH, 45229, USA
| | - Michelle Sargent
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Mike Adam
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Qing Ma
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, 268-280 Albert Sabin Way, location T, floor 6, suite 272, Cincinnati, OH, 45229, USA
| | - Anne Karina T Perl
- Division of Neonatology and Pulmonary biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Alexander G Miethke
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ashley Burg
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tiffany Shi
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - David A Hildeman
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - E Steve S Woodle
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - J Matthew Kofron
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Prasad Devarajan
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, 268-280 Albert Sabin Way, location T, floor 6, suite 272, Cincinnati, OH, 45229, USA.
| |
Collapse
|
12
|
Li X, Li N, Wang Y, Han Q, Sun B. Research Progress of Fibroblasts in Human Diseases. Biomolecules 2024; 14:1478. [PMID: 39595654 PMCID: PMC11591654 DOI: 10.3390/biom14111478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Fibroblasts, which originate from embryonic mesenchymal cells, are the predominant cell type seen in loose connective tissue. As the main components of the internal environment that cells depend on for survival, fibroblasts play an essential role in tissue development, wound healing, and the maintenance of tissue homeostasis. Furthermore, fibroblasts are also involved in several pathological processes, such as fibrosis, cancers, and some inflammatory diseases. In this review, we analyze the latest research progress on fibroblasts, summarize the biological characteristics and physiological functions of fibroblasts, and delve into the role of fibroblasts in disease pathogenesis and explore treatment approaches for fibroblast-related diseases.
Collapse
Affiliation(s)
| | | | | | | | - Boshi Sun
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; (X.L.); (N.L.); (Y.W.); (Q.H.)
| |
Collapse
|