1
|
Yuan J, Deng T, Yang Q, Lv D, Zhou Z, You L, Feng Q, Meng X, Pang Q, Li H, Zhu B. Loss of LSD1 ameliorates myocardial infarction by regulating angiogenesis via transcriptional activation of Vegfa. Life Sci 2025; 372:123613. [PMID: 40210117 DOI: 10.1016/j.lfs.2025.123613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/13/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025]
Abstract
AIMS Our study aims to explore the regulatory role and underlying mechanisms of Lysine-specific demethylase 1 (LSD1) in angiogenesis following myocardial infarction (MI). MATERIALS AND METHODS We generated inducible cardiomyocyte-specific Lsd1 knockout (Lsd1-cKO) mice and established a MI model. The function of LSD1 in cardiac angiogenesis in MI mice was assessed through echocardiography, histopathological staining, and immunofluorescence analysis. In vitro, Lsd1 silencing in cardiomyocytes was achieved by transfecting small interfering RNA (siRNA), followed by hypoxic treatment to simulate the in vivo MI model. The above cardiomyocyte-conditioned medium was collected and used to treat endothelial cells to observe changes in endothelial function. Additionally, we employed Cleavage Under Targets and Tagmentation sequencing (CUT&Tag-seq) to investigate the potential mechanisms by which LSD1 exerts its effects. KEY FINDINGS We found that the absence of LSD1 protected against cardiac dysfunction and promoted angiogenesis in mice with MI. Lsd1-silenced cardiomyocytes enhance the migration and tube formation function of endothelial cells by releasing vascular endothelial growth factor A (VEGF-A) under hypoxic conditions. The combined analysis of CUT&Tag-seq data revealed that silencing of Lsd1 promoted the monomethylation of H3K4 at the Vegfa promoter and region, leading to the transcriptional activation of Vegfa mRNA in cardiomyocytes. SIGNIFICANCE Our research indicates that lowered level of LSD1 in cardiomyocytes enhances VEGF-A paracrine secretion and improves endothelial cell function through cross-talk, ultimately promoting angiogenesis. These findings suggest that targeting LSD1 might be an effective therapeutic approach to protect against MI.
Collapse
Affiliation(s)
- Jinghan Yuan
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tian Deng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qingshan Yang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Danyi Lv
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhenfang Zhou
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lu You
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qipu Feng
- Animal Experiment Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Xiangmin Meng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiuyu Pang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hao Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bingmei Zhu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
2
|
Wang Y, Liang Y, Li M, Lu J, Zhou S, Yu Y, Yang C, Hou X. Single-cell multi-omics reveals that FABP1 + renal cell carcinoma drive tumor angiogenesis through the PLG-PLAT axis under fatty acid reprogramming. Mol Cancer 2025; 24:179. [PMID: 40518526 DOI: 10.1186/s12943-025-02377-9] [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: 03/21/2025] [Accepted: 06/03/2025] [Indexed: 06/18/2025] Open
Abstract
Renal cell carcinoma is characterized by a poor prognosis. Recently, renal cell carcinoma has been recognized as a metabolic disease associated with fatty acid metabolic reprogramming, although in-depth studies on this topic are still lacking. We found that fatty acid metabolism reprogramming in renal cell carcinoma is primarily characterized by high expression of FABP1. FABP1 + tumors significantly impact survival and display distinct differentiation trajectories compared to other tumor subclusters. They show elevated expression of angiogenesis and cell migration signals, with PLG-PLAT-mediated interactions with endothelial cells notably enhanced. Spatial transcriptomics show a prominent co-localization of FABP1 + tumors with endothelial cells, and their spatial distribution closely aligns with that of PLAT + endothelial cells. FABP1 + tumors exhibit a unique pattern in spatial transcriptomics, enriched in Extracellular Matrix and angiogenesis-related pathways. Through receptor-ligand interaction analysis, a novel PLG-PLAT functional axis was found between tumor epithelial cells and endothelial cells. Based on results of experiments, we infer that FABP1 + tumors can promote plasmin-related tumor angiogenesis by triggering the PLG-PLAT signaling axis. Finally, utilizing preclinical models, we suggest that targeting the FABP1-PLG-PLAT axis may serve as promising strategy enhancing the sensitivity of Tyrosine Kinase Inhibitor therapy.
Collapse
Affiliation(s)
- Yiqiu Wang
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yingchun Liang
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Min Li
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Jiayi Lu
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Sian Zhou
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yaoyu Yu
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Changwei Yang
- Department of Nutrition and Food Safety, School of Public Health, Fujian Medical University, Fuzhou, 350122, China.
| | - Xinhuang Hou
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Vascular Surgery, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
3
|
Liu C, Zhang X, Xie Q, Fang B, Liu F, Luo J, Aihemaiti G, Ji W, Yang Y, Li X. Identification of key proteins and pathways in myocardial infarction using machine learning approaches. Sci Rep 2025; 15:19530. [PMID: 40467768 PMCID: PMC12137932 DOI: 10.1038/s41598-025-04401-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 05/27/2025] [Indexed: 06/11/2025] Open
Abstract
Acute myocardial infarction (AMI) is a leading cause of global morbidity and mortality, requiring deeper insights into its molecular mechanisms for improved diagnosis and treatment. This study combines proteomics, transcriptomics and machine learning (ML) to identify key proteins and pathways associated with AMI. Plasma samples from 48 AMI patients and 50 healthy controls (HC) were used for proteomic sequencing. Differentially expressed proteins (DEPs) were identified and analyzed for pathway enrichment. Protein-protein interaction (PPI) networks were constructed, and we conducted a meta-analysis (GSE60993, GSE61144, GSE48060) using an inverse variance model to combine differentially expressed genes (DEGs) identified via LIMMA and FDR adjustment across three studies. Clustering and co-expression analysis were performed using K-Medoids and weighted gene co-expression network analysis (WGCNA). ML feature selection identified hub proteins, which were validated across bulk, single-cell, and spatial datasets for atherosclerosis (ATH) and MI. In this study, we identified 437 DEPs with 291 up-regulated and 146 down-regulated proteins. Functional enrichment analysis revealed key pathways involved in inflammation, immunity, metabolism, and cellular stress responses, among others. Using non-negative matrix factorization (NNMF) and K-Medoids clustering, AMI patients were divided into two clusters (C1 and C2), with distinct protein expression patterns and inflammatory responses. Differential analysis between clusters revealed 200 cluster-specific DEPs, with C1 associated with angiogenesis and vascular remodeling, and C2 linked to cellular stress and apoptosis. A meta-analysis identified 1383 DEGs, and their intersection with DEPs yielded 63 proteins, which were subsequently refined by logistic regression to 36 AMI-associated proteins. Furthermore, a protein co-expression network analysis identified 49 modules, with the turquoise module being strongly associated with AMI highlighting pathways in lipid metabolism, immune response, and tissue repair. From this module, 17 key proteins were selected, and ML further distilled these to nine core features (CAMP, CLTC, CTNNB1, FUBP3, IQGAP1, MANBA, ORM1, PSME1, and SPP1) that are closely linked to immune regulation, apoptosis, and metabolism. These proteins were validated across multiple datasets. Single-cell analysis revealed distinct expression patterns of these proteins across cell types and spatial regions in ATH and MI, emphasizing their roles in inflammation, vascular remodeling, and plaque instability. This study identifies critical proteins and pathways in AMI, offering potential biomarkers and therapeutic targets. The use of ML provides a robust framework for identifying AMI's key molecular.
Collapse
Affiliation(s)
- Chang Liu
- Department of Cardiology, The first Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Xing Zhang
- Department of Cardiology, The first Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Qian Xie
- Department of Cardiology, The first Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Binbin Fang
- Department of Cardiology, The first Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Fen Liu
- Department of Cardiology, The first Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Junyi Luo
- Department of Cardiology, The first Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Gulandanmu Aihemaiti
- Department of Cardiology, The first Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Wei Ji
- Department of Cardiology, The first Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Yining Yang
- Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, 830001, China.
| | - Xiaomei Li
- Department of Cardiology, The first Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China.
| |
Collapse
|
4
|
Yang S, Penna V, Lavine KJ. Functional diversity of cardiac macrophages in health and disease. Nat Rev Cardiol 2025; 22:431-442. [PMID: 39743564 DOI: 10.1038/s41569-024-01109-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/04/2025]
Abstract
Macrophages make up a substantial portion of the stromal compartment of the heart in health and disease. In the past decade, the origins of these cardiac macrophages have been established as two broad populations derived from either embryonic or definitive haematopoiesis and that can be distinguished by the expression of CC-motif chemokine receptor 2 (CCR2). These cardiac macrophage populations are transcriptionally distinct and have differing cell surface markers and divergent roles in cardiac homeostasis and disease. Embryonic-derived CCR2- macrophages are a tissue-resident population that participates in tissue development, repair and maintenance, whereas CCR2+ macrophages are derived from definitive haematopoiesis and contribute to inflammation and tissue damage. Studies from the past 5 years have leveraged single-cell RNA sequencing technologies to expand our understanding of cardiac macrophage diversity, particularly of the monocyte-derived macrophage populations that reside in the injured and diseased heart. Emerging technologies in spatial transcriptomics have enabled the identification of distinct disease-associated cellular neighbourhoods consisting of macrophages, other immune cells and fibroblasts, highlighting the involvement of macrophages in cell-cell communication. Together, these discoveries lend new insights into the role of specific macrophage populations in the pathogenesis of cardiac disease, which can pave the way for the identification of new therapeutic targets and the development of diagnostic tools. In this Review, we discuss the developmental origin of cardiac macrophages and describe newly identified cell states and associated cellular neighbourhoods in the steady state and injury settings. We also discuss various contributions and effector functions of cardiac macrophages in homeostasis and disease.
Collapse
Affiliation(s)
- Steven Yang
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Vinay Penna
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kory J Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
5
|
Xu C, Su R, Song Y, Shu W, Yang M, Yang Z, Xu X, Wei X. Integrating spatial transcriptomics and single-cell RNA-seq dissects immune microenvironment in fatty liver regeneration. Clin Transl Med 2025; 15:e70365. [PMID: 40515443 PMCID: PMC12166122 DOI: 10.1002/ctm2.70365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/23/2025] [Accepted: 05/28/2025] [Indexed: 06/16/2025] Open
Affiliation(s)
- Chenhao Xu
- Department of Hepatobiliary and Pancreatic SurgeryHangzhou First People's Hospital, Zhejiang University School of MedicineHangzhouChina
- Institute of Translational Medicine, Zhejiang University School of MedicineHangzhouChina
| | - Renyi Su
- Department of Hepatobiliary and Pancreatic SurgeryHangzhou First People's Hospital, Zhejiang University School of MedicineHangzhouChina
- Institute of Translational Medicine, Zhejiang University School of MedicineHangzhouChina
| | - Yisu Song
- Department of Hepatobiliary and Pancreatic SurgeryHangzhou First People's Hospital, Zhejiang University School of MedicineHangzhouChina
- Institute of Translational Medicine, Zhejiang University School of MedicineHangzhouChina
| | - Wenzhi Shu
- Department of Hepatobiliary and Pancreatic SurgeryHangzhou First People's Hospital, Zhejiang University School of MedicineHangzhouChina
- Institute of Translational Medicine, Zhejiang University School of MedicineHangzhouChina
| | - Mengfan Yang
- Department of Organ TransplantationQilu Hospital of Shandong UniversityJinanChina
| | - Zhe Yang
- Department of Hepatobiliary and Pancreatic SurgeryDepartment of Liver TransplantationShulan (Hangzhou) Hospital, Zhejiang Shuren University School of MedicineHangzhouChina
| | - Xiao Xu
- Institute of Translational Medicine, Zhejiang University School of MedicineHangzhouChina
- School of Clinical Medicine, Hangzhou Medical CollegeHangzhouChina
- NHC Key Laboratory of Combined Multi‐organ TransplantationHangzhouChina
| | - Xuyong Wei
- Department of Hepatobiliary and Pancreatic SurgeryHangzhou First People's Hospital, Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
6
|
Gomez-Salinero JM, Redmond D, Rafii S. Microenvironmental determinants of endothelial cell heterogeneity. Nat Rev Mol Cell Biol 2025; 26:476-495. [PMID: 39875728 DOI: 10.1038/s41580-024-00825-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 01/30/2025]
Abstract
During development, endothelial cells (ECs) undergo an extraordinary specialization by which generic capillary microcirculatory networks spanning from arteries to veins transform into patterned organotypic zonated blood vessels. These capillary ECs become specialized to support the cellular and metabolic demands of each specific organ, including supplying tissue-specific angiocrine factors that orchestrate organ development, maintenance of organ-specific functions and regeneration of injured adult organs. Here, we illustrate the mechanisms by which microenvironmental signals emanating from non-vascular niche cells induce generic ECs to acquire specific inter-organ and intra-organ functional attributes. We describe how perivascular, parenchymal and immune cells dictate vascular heterogeneity and capillary zonation, and how this system is maintained through tissue-specific signalling activated by vasculogenic and angiogenic factors and deposition of matrix components. We also discuss how perturbation of organotypic vascular niche cues lead to erasure of EC signatures, contributing to the pathogenesis of disease processes. We also describe approaches that use reconstitution of tissue-specific signatures of ECs to promote regeneration of damaged organs.
Collapse
Affiliation(s)
- Jesus M Gomez-Salinero
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - David Redmond
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Wang K, Li M, Sartor MA, Colacino JA, Dolinoy DC, Svoboda LK. Perinatal Exposure to Lead or Diethylhexyl Phthalate in Mice: Sex-Specific Effects on Cardiac DNA Methylation and Gene Expression across Time. ENVIRONMENTAL HEALTH PERSPECTIVES 2025; 133:67014. [PMID: 40315424 DOI: 10.1289/ehp15503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
Abstract
BACKGROUND Global and site-specific changes in DNA methylation and gene expression are associated with cardiovascular development, aging, and disease, but how the transcriptome and epigenome of the heart change across the life course in males vs. females and how chemical exposures early in life influence this programming have not yet been investigated. OBJECTIVES We used an established mouse model of developmental exposures to investigate the effects of perinatal exposure to either lead (Pb) or diethylhexyl phthalate (DEHP), two ubiquitous environmental contaminants that are both strongly associated with cardiovascular diseases (CVDs), on DNA methylation and gene expression across the life course in whole hearts. METHODS Dams were randomly assigned to receive human physiologically relevant levels of Pb (32 ppm in water), DEHP (25 mg / kg chow), or control water and chow. Exposures started 2 weeks prior to mating and continued until weaning at postnatal day 21 (3 wk of age). Approximately 1 male and 1 female offspring per litter were followed to 3 wk, 5 months, or 10 months of age, at which time whole hearts were collected (n ≥ 5 per sex per exposure). Enhanced reduced representation bisulfite sequencing (ERRBS) was used to assess the cardiac DNA methylome at 3 wk and 10 months, and RNA-Seq was conducted at all three time points. MethylSig and edgeR were used to identify age-related differentially methylated regions (DMRs) and differentially expressed genes (DEGs), respectively, within each sex and exposure group. Cell type deconvolution of bulk RNA-Seq data was conducted using the MuSiC algorithm and publicly available single-cell RNA-Seq data. RESULTS Thousands of DMRs and hundreds of DEGs were identified in control, DEHP, and Pb-exposed hearts across time between 3 wk and 10 months of age. A closer look at the genes and pathways showing differential DNA methylation revealed that the majority were unique to each sex and exposure group. Overall, pathways governing development and differentiation changed across time in all conditions. A small number of genes in each group showed significant differences in DNA methylation and gene expression with life stage, including several that were different in toxicant-exposed but not control mice. We also observed subtle but significant differences in the proportion of several cell types that were associated with life stage, sex, or developmental exposure. DISCUSSION Together these data suggest that gene expression and DNA methylation programs, as well as cellular composition, may differ across the life course long after cessation of exposure in perinatal Pb- or DEHP-exposed mice compared to controls and highlight potential biomarkers of developmental toxicant exposures; however, additional studies are required for confirmation. Further studies are also needed to investigate how epigenetic and transcriptional differences impact cardiovascular health across the life course, particularly in old age when the risk of cardiovascular diseases is markedly increased. https://doi.org/10.1289/EHP15503.
Collapse
Affiliation(s)
- Kai Wang
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, Michigan, USA
| | - Minghua Li
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, Michigan, USA
| | - Maureen A Sartor
- Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Justin A Colacino
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Dana C Dolinoy
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Laurie K Svoboda
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
8
|
Yang C, Li H, Chen Y, Zhu W, Wang J. Omics-based Approach Towards Macrophages: New Perspectives of Biology and Function in the Normal and Diseased Heart. Int J Biol Sci 2025; 21:3666-3688. [PMID: 40520014 PMCID: PMC12160929 DOI: 10.7150/ijbs.112061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 05/07/2025] [Indexed: 06/18/2025] Open
Abstract
Macrophages play a crucial role not only in maintaining homeostasis but also in initiating inflammatory responses to various forms of stress or injury, thereby contributing to tissue damage while concurrently promoting recovery. Furthermore, the diversity of macrophage subtypes, their spatial distribution, and distinct cellular functions are closely linked to the pathogenesis and severity of cardiovascular diseases such as myocardial infarction, atherosclerosis, heart failure, and myocarditis. This association underscores the importance of investigating macrophage heterogeneity in different pathological contexts. Recent advances in multi-omics technologies-including single-cell RNA sequencing, spatial transcriptomics, and metabolomics-have elucidated the heterogeneity of macrophages, their intercellular interactions, underlying functional mechanisms, and spatial organization. In this review, we systematically summarize the diverse phenotypes and functional plasticity of macrophages in the regulation of cardiovascular diseases, with particular emphasis on the novel insights afforded by multi-omics approaches. We focus on the characteristics of macrophages in both physiological and pathological states, thereby providing reference points for clinical macrophage-targeted strategies and their translational significance.
Collapse
Affiliation(s)
- Chao Yang
- Department of Cardiology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
- Transvascular Implantation Devices Research Institute (TIDRI), Hangzhou 310053, China
| | - Huajun Li
- Department of Cardiology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yuxing Chen
- Department of Cardiology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang province, Hangzhou 310009, China
| | - Wei Zhu
- Department of Cardiology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang province, Hangzhou 310009, China
| | - Jian'an Wang
- Department of Cardiology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang province, Hangzhou 310009, China
- Transvascular Implantation Devices Research Institute (TIDRI), Hangzhou 310053, China
| |
Collapse
|
9
|
Xu Z, Wang Y, Xie T, Luo R, Ni HL, Xiang H, Tang S, Tan S, Fang R, Ran P, Zhang Q, Xu X, Tian S, He F, Yang W, Ding C. Panoramic spatial enhanced resolution proteomics (PSERP) reveals tumor architecture and heterogeneity in gliomas. J Hematol Oncol 2025; 18:58. [PMID: 40420200 DOI: 10.1186/s13045-025-01710-5] [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: 09/04/2024] [Accepted: 04/29/2025] [Indexed: 05/28/2025] Open
Abstract
The spatial proteomic profiling of complex tissues is essential for investigating cellular function in physiological and pathological states. However, the imbalance among resolution, protein coverage, and expense precludes their systematic application to analyze whole tissue sections in an unbiased manner and with high resolution. Here, we introduce panoramic spatial enhanced resolution proteomics (PSERP), a method that combines tissue expansion, automated sample segmentation, and tryptic digestion with high-throughput proteomic profiling. The PSERP approach facilitates rapid quantitative profiling of proteomic spatial variability in whole tissue sections at sub-millimeter resolution. We demonstrated the utility of this method for determining the streamlined large-scale spatial proteomic features of gliomas. Specifically, we profiled spatial proteomic features for nine glioma samples across three different mutation types (IDH1-WT/EGFR-mutant, IDH1-mutant, and IDH1/EGFR-double-WT gliomas) at sub-millimeter resolution (corresponding to a total of 2,230 voxels). The results revealed over 10,000 proteins identified in a single slide, which helps us to portray the diverse proteins and pathways with spatial abundance patterns in the context of tumor heterogeneity and cellular features. Our spatial proteomic data revealed distinctive proteomic features of malignant and non-malignant tumor regions and depicted the distribution of proteins from tumor centers to tumor borders and non-malignant tumor regions. Through integrative analysis with single-cell transcriptomic data, we elucidated the cellular composition and cell-cell communications in a spatial context. Our PSERP also includes a spatially resolved tumor-specific peptidome identification workflow that not only enables us to elucidate the spatial expression patterns of tumor-specific peptides in glioma samples with different genomic types but also provides us with opportunities to select combinations of tumor-specific mutational peptides whose expression could cover the maximum tumor regions for future immune therapies. We further demonstrated that combining tumor-specific peptides might enhance the efficacy of immunotherapy in both patient-derived cell (PDC) and patient-derived xenograft (PDX) models. PSERP efficiently retains precise spatial proteomic information within the tissue context and provides a deeper understanding of tissue biology and pathology at the molecular level.
Collapse
Affiliation(s)
- Ziyan Xu
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yunzhi Wang
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Tao Xie
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rongkui Luo
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Heng-Li Ni
- Department of Pathology, Children's Hospital of Soochow University, Soochow University, Suzhou, JiangSu Province, 215000, The People's Republic of China
| | - Hang Xiang
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Shaoshuai Tang
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Subei Tan
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Rundong Fang
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Peng Ran
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Qiao Zhang
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiaomeng Xu
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Sha Tian
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Fuchu He
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing, China
- Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Wenjun Yang
- Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China.
| | - Chen Ding
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- Departments of Cancer Research Institute, Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Translational Biomedical Engineering, Urumqi 830000, China.
| |
Collapse
|
10
|
Mohanta SK, Heron C, Klaus-Bergmann A, Horstmann H, Brakenhielm E, Giannarelli C, Habenicht AJR, Gerhardt H, Weber C. Metabolic and Immune Crosstalk in Cardiovascular Disease. Circ Res 2025; 136:1433-1453. [PMID: 40403115 DOI: 10.1161/circresaha.125.325496] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
Cardiovascular diseases including atherosclerosis and heart failure, arise from the intricate interplay of metabolic, immune, and neural dysregulation within vascular and cardiac tissues: This review focuses on integrating recent advances in metabolic and immune crosstalk of the cardiac vasculature that affects cardiometabolic health and disease progression. Coronary and lymphatic endothelial cells regulate cardiac metabolism, and their dysfunction is linked to cardiovascular diseases. Lymphatics maintain tissue homeostasis, including clearing metabolic waste, lipids, and immune cells, and their maladaptation in metabolic diseases worsens outcomes. Altered vascular endothelial metabolism in heart failure drives immune-mediated inflammation, fibrosis, and adverse cardiac remodeling. Concurrently, artery tertiary lymphoid organs formed in the adventitia of advanced atherosclerotic arteries, serve as pivotal neuroimmune hubs, coordinating local immunity through T and B cell activation and neurovascular signaling via artery-brain circuits. T cells within plaques and artery tertiary lymphoid organs undergo clonal expansion as a result of peripheral tolerance breakdown, with proinflammatory CD4+ and CD8+ subsets amplifying atherosclerosis, effects further shaped by systemic immune activation. Therapeutic strategies targeting endothelial cell metabolism, lymphatic dysfunction, neuroimmune crosstalk, and T cell plasticity hold promise for integrated cardiovascular disease management.
Collapse
Affiliation(s)
- Sarajo K Mohanta
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU), LMU University Hospital, Munich, Germany (S.K.M., A.J.R.H., C.W.)
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (S.K.M., A.J.R.H., C.W.)
| | - Coraline Heron
- UnivRouen Normandie, INSERM EnVI, UMR 1096, Rouen, France (C.H., E.B.)
| | - Alexandra Klaus-Bergmann
- Integrative Vascular Biology Laboratory, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (A.K.-B., H.G.)
- German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany (A.K.-B., H.G.)
| | - Hauke Horstmann
- Cardiology and Angiology, Medical Center (H.H.), University of Freiburg, Freiburg, Germany
- Faculty of Medicine (H.H.), University of Freiburg, Freiburg, Germany
- Department of Medicine, Division of Cardiology (H.H., C.G.), NYU Grossman School of Medicine, New York, NY
| | - Ebba Brakenhielm
- UnivRouen Normandie, INSERM EnVI, UMR 1096, Rouen, France (C.H., E.B.)
| | - Chiara Giannarelli
- Department of Medicine, Division of Cardiology (H.H., C.G.), NYU Grossman School of Medicine, New York, NY
- Department of Pathology (C.G.), NYU Grossman School of Medicine, New York, NY
| | - Andreas J R Habenicht
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU), LMU University Hospital, Munich, Germany (S.K.M., A.J.R.H., C.W.)
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (S.K.M., A.J.R.H., C.W.)
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China (A.J.R.H.)
| | - Holger Gerhardt
- Integrative Vascular Biology Laboratory, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (A.K.-B., H.G.)
- German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany (A.K.-B., H.G.)
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU), LMU University Hospital, Munich, Germany (S.K.M., A.J.R.H., C.W.)
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (S.K.M., A.J.R.H., C.W.)
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands (C.W.)
| |
Collapse
|
11
|
Wang X, Chen L, Wei J, Zheng H, Zhou N, Xu X, Deng X, Liu T, Zou Y. The immune system in cardiovascular diseases: from basic mechanisms to therapeutic implications. Signal Transduct Target Ther 2025; 10:166. [PMID: 40404619 PMCID: PMC12098830 DOI: 10.1038/s41392-025-02220-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/22/2024] [Accepted: 03/20/2025] [Indexed: 05/24/2025] Open
Abstract
Immune system plays a crucial role in the physiological and pathological regulation of the cardiovascular system. The exploration history and milestones of immune system in cardiovascular diseases (CVDs) have evolved from the initial discovery of chronic inflammation in atherosclerosis to large-scale clinical studies confirming the importance of anti-inflammatory therapy in treating CVDs. This progress has been facilitated by advancements in various technological approaches, including multi-omics analysis (single-cell sequencing, spatial transcriptome et al.) and significant improvements in immunotherapy techniques such as chimeric antigen receptor (CAR)-T cell therapy. Both innate and adaptive immunity holds a pivotal role in CVDs, involving Toll-like receptor (TLR) signaling pathway, nucleotide-binding oligomerization domain-containing proteins 1 and 2 (NOD1/2) signaling pathway, inflammasome signaling pathway, RNA and DNA sensing signaling pathway, as well as antibody-mediated and complement-dependent systems. Meanwhile, immune responses are simultaneously regulated by multi-level regulations in CVDs, including epigenetics (DNA, RNA, protein) and other key signaling pathways in CVDs, interactions among immune cells, and interactions between immune and cardiac or vascular cells. Remarkably, based on the progress in basic research on immune responses in the cardiovascular system, significant advancements have also been made in pre-clinical and clinical studies of immunotherapy. This review provides an overview of the role of immune system in the cardiovascular system, providing in-depth insights into the physiological and pathological regulation of immune responses in various CVDs, highlighting the impact of multi-level regulation of immune responses in CVDs. Finally, we also discuss pre-clinical and clinical strategies targeting the immune system and translational implications in CVDs.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Liming Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianming Wei
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Hao Zheng
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Ning Zhou
- Department of Cardiovascular Medicine, Anzhen Hospital Affiliated to Capital Medical University, Beijing, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Deng
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tao Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China.
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Jiangsu, Nanjing, China.
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
- Institutes of Advanced Medical Sciences and Huaihe Hospital, Henan University, Kaifeng, Henan, China.
| |
Collapse
|
12
|
Fließer E, Jandl K, Chen SH, Wang MT, Schupp JC, Kuebler WM, Baker AH, Kwapiszewska G. Transcriptional signatures of endothelial cells shape immune responses in cardiopulmonary health and disease. JCI Insight 2025; 10:e191059. [PMID: 40401523 DOI: 10.1172/jci.insight.191059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025] Open
Abstract
The cardiopulmonary vasculature and its associated endothelial cells (ECs) play an essential role in sustaining life by ensuring the delivery of oxygen and nutrients. Beyond these foundational functions, ECs serve as key regulators of immune responses. Recent advances in single-cell RNA sequencing have revealed that the cardiopulmonary vasculature is composed of diverse EC subpopulations, some of which exhibit specialized immunomodulatory properties. Evidence for immunomodulation includes distinct expression profiles associated with antigen presentation, cytokine secretion, immune cell recruitment, translocation, and clearance - functions critical for maintaining homeostasis in the heart and lungs. In cardiopulmonary diseases, ECs undergo substantial transcriptional reprogramming, leading to a shift from homeostasis to an activated state marked by heightened immunomodulatory activity. This transformation has highlighted the critical role for ECs in disease pathogenesis and their potential as future therapy targets. This Review emphasizes the diverse functions of ECs in the heart and lungs, particularly adaptive and maladaptive immunoregulatory roles in cardiopulmonary health and disease.
Collapse
Affiliation(s)
- Elisabeth Fließer
- Otto Loewi Research Center, Lung Research Cluster, Medical University of Graz, Graz, Austria
- Institute for Lung Health, Cardiopulmonary Institute, Member of German Lung Center, Justus-Liebig University, Giessen, Germany
| | - Katharina Jandl
- Otto Loewi Research Center, Lung Research Cluster, Medical University of Graz, Graz, Austria
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Shiau-Haln Chen
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Mei-Tzu Wang
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jonas C Schupp
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pulmonary and Infectious Diseases, Hannover Medical School, Hannover, Germany
- Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), German Center for Lung Research BREATH, Hannover, Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin, Berlin, Germany
- German Center for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
- German Center for Lung Research, Associated Partner Site Berlin, Berlin, Germany
- Department of Surgery and
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre, St Michael's Hospital, Toronto, Ontario, Canada
| | - Andrew H Baker
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Department of Pathology, Cardiovascular Research Institute Maastricht, School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
| | - Grazyna Kwapiszewska
- Otto Loewi Research Center, Lung Research Cluster, Medical University of Graz, Graz, Austria
- Institute for Lung Health, Cardiopulmonary Institute, Member of German Lung Center, Justus-Liebig University, Giessen, Germany
| |
Collapse
|
13
|
Li Z, Rius Rigau A, Xie W, Huang L, Ye W, Li YN, Matei AE, Bergmann C, Shao X, Zou H, Wang J, Pinello L, Distler JHW, He R, Liang M. Spatial multiomics decipher fibroblast-macrophage dynamics in systemic sclerosis. Ann Rheum Dis 2025:S0003-4967(25)00953-7. [PMID: 40410053 DOI: 10.1016/j.ard.2025.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/18/2025] [Accepted: 04/27/2025] [Indexed: 05/25/2025]
Abstract
OBJECTIVES Stromal-immune crosstalk shapes the pathogenic microenvironment of systemic sclerosis (SSc), but the spatial regulatory networks underlying fibrogenesis remain poorly defined. We aimed to explore tissue organisation and cell coordination in SSc skin, providing spatiotemporal insights into disease mechanisms and bridging the gap between omics discovery and precision medicine. METHODS We performed spatial transcriptomics on skin biopsies from 10 patients with diffuse cutaneous SSc and 4 healthy controls using the 10× Visium platform. These findings were confirmed using higher-resolution Stereo-seq transcriptomics, spatial proteomics, and single-cell RNA sequencing data from patients with SSc, SSc mouse models, and wound-healing reindeer models. In vivo and in vitro studies were conducted to validate the key regulatory pathways. RESULTS Fourteen skin biopsies were analysed, revealing significant expansion of fibrotic niches enriched with fibroblasts and macrophages in SSc, correlating with clinical severity. We revealed disease-specific cell states of fibroblasts and macrophages and evaluated their spatial dependency and cell-cell communication. Stratification based on signature genes enabled the identification of patients with SSc with progressive disease and treatment-nonresponsive phenotype. ACKR3 (a CXCL12 decoy receptor) was selectively expressed in myofibroblast progenitors, which diminished during differentiation towards mature myofibroblast, potentially serving to regulate CXCL12/CXCR4-mediated proinflammatory macrophage recruitment. Inhibition of CXCR4 attenuated skin and lung fibrosis in experimental fibrosis mouse models. CONCLUSIONS Our spatially resolved atlas uncovered dynamic fibroblast-macrophage interplay as a hallmark of fibrotic niche expansion. These findings offer spatiotemporal insights into disease mechanisms and pave the way for advanced mechanistic and therapeutic studies, bridging the gap between omics discovery and precision medicine.
Collapse
Affiliation(s)
- Zhijian Li
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Molecular Pathology Unit, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Aleix Rius Rigau
- Department of Internal Medicine 3, Rheumatology and Clinical Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Wenjie Xie
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Linlin Huang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China; Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenjing Ye
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China; Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi-Nan Li
- Clinic for Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany; Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany
| | - Alexandru-Emil Matei
- Clinic for Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany; Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany
| | - Christina Bergmann
- Department of Internal Medicine 3, Rheumatology and Clinical Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Xiaohang Shao
- Department of Nephrology and Rheumatology Immunology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Hejian Zou
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China; Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiucun Wang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China; State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai, China; Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Shanghai, China
| | - Luca Pinello
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Molecular Pathology Unit, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Jörg H W Distler
- Clinic for Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany; Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany.
| | - Rui He
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| | - Minrui Liang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China; Huashan Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
14
|
Dai Z, Ding H, Zhang Q, Fu L, Tai S. Spatial Insights in Cardiovascular Aging. Aging Dis 2025:AD.2025.0272. [PMID: 40423633 DOI: 10.14336/ad.2025.0272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 04/28/2025] [Indexed: 05/28/2025] Open
Abstract
Spatial omics provides unprecedented insights into how the cardiovascular system is spatially organized and how cellular phenotypes are distributed. Researchers have been able to clarify the complex spatial architecture of the cardiovascular system and how cellular phenotypes are distributed during the aging process by integrating data from spatial omics. In addition, this new technology has revealed previously hidden patterns of gene expression and cellular communication that were not detected using traditional bulk omics approaches. In this review, we explore the contribution of spatial omics in clarifying the molecular mechanisms that influence cardiovascular aging, highlighting the importance and application of spatial omics in unraveling the spatial heterogeneity within the aging cardiovascular system. This will help us understand the molecular mechanisms implicated in age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Zhongling Dai
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Huiqin Ding
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Quan Zhang
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Liyao Fu
- Department of Blood Transfusion, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shi Tai
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| |
Collapse
|
15
|
Dupuis L, Debeaupuis O, Simon F, Isambert H. CausalCCC: a web server to explore intracellular causal pathways enabling cell-cell communication. Nucleic Acids Res 2025:gkaf404. [PMID: 40366019 DOI: 10.1093/nar/gkaf404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/26/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
Understanding cell-cell communication (CCC) pathways from single-cell or spatial transcriptomic data is key to unraveling biological processes. Recently, multiple CCC methods have been developed but primarily focus on refining ligand-receptor (L-R) interaction scores. A critical gap for a more comprehensive picture of cellular crosstalks lies in the integration of upstream and downstream intracellular pathways in the sender and receiver cells. We report here CausalCCC, https://miic.curie.fr/causalCCC.php, an interactive web server, which addresses this need by reconstructing gene-gene interaction pathways across two or more interacting cell types from single-cell or spatial transcriptomic data. CausalCCC includes a graphical introduction and a demo dataset within the workbench page as well as a comprehensive tutorial. CausalCCC methodology integrates a robust and scalable causal network reconstruction method, multivariate information-based inductive causation, with internally computed L-R pairs using LIANA+ (including CellphoneDBv5, SingleCellSignalR, Connectome, NATMI, and Log2FC). Alternatively, user-defined L-R pairs from any CCC methods can also be uploaded. We showcase here CausalCCC on different single-cell and spatial transcriptomic datasets from three original CCC methods (NicheNet, CellChat, and Misty). CausalCCC web server offers unique interactive visualization tools dedicated to single-cell data practitioners seeking to go beyond L-R scores and explore extended CCC pathways across multiple interacting cell types.
Collapse
Affiliation(s)
| | - Orianne Debeaupuis
- CNRS UMR168, Institut Curie, 75005 Paris, France
- Inserm U1163, Institut Imagine, 75005 Paris, France
| | - Franck Simon
- CNRS UMR168, Institut Curie, 75005 Paris, France
| | | |
Collapse
|
16
|
Garitano N, Aguado-Alvaro LP, Pelacho B. Emerging Epigenetic Therapies for the Treatment of Cardiac Fibrosis. Biomedicines 2025; 13:1170. [PMID: 40426997 PMCID: PMC12109272 DOI: 10.3390/biomedicines13051170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/27/2025] [Accepted: 05/03/2025] [Indexed: 05/29/2025] Open
Abstract
Fibrosis is a pathological process characterized by excessive extracellular matrix (ECM) deposition, leading to tissue stiffening and organ dysfunction. It is a major contributor to chronic diseases affecting various organs, with limited therapeutic options available. Among the different forms of fibrosis, cardiac fibrosis is particularly relevant due to its impact on cardiovascular diseases (CVDs), which remain the leading cause of morbidity and mortality worldwide. This process is driven by activated cardiac fibroblasts (CFs), which promote ECM accumulation in response to chronic stressors. Epigenetic mechanisms, including DNA methylation, histone modifications, and chromatin remodeling, are key regulators of fibroblast activation and fibrotic gene expression. Enzymes such as DNA methyltransferases (DNMTs), histone methyltransferases (HMTs), histone acetyltransferases (HATs), and histone deacetylases (HDACs) have emerged as potential therapeutic targets, and epigenetic inhibitors have shown promise in modulating these enzymes to attenuate fibrosis by controlling fibroblast function and ECM deposition. These small-molecule compounds offer advantages such as reversibility and precise temporal control, making them attractive candidates for therapeutic intervention. This review aims to provide a comprehensive overview of the mechanisms by which epigenetic regulators influence cardiac fibrosis and examines the latest advances in preclinical epigenetic therapies. By integrating recent data from functional studies, single-cell profiling, and drug development, it highlights key molecular targets, emerging therapeutic strategies, and current limitations, offering a critical framework to guide future research and clinical translation.
Collapse
Affiliation(s)
- Nerea Garitano
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (N.G.); (L.P.A.-A.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Laura Pilar Aguado-Alvaro
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (N.G.); (L.P.A.-A.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Beatriz Pelacho
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (N.G.); (L.P.A.-A.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| |
Collapse
|
17
|
Xie Y, Tucciarone L, Farah EN, Chang L, Yang Q, Shankar TS, Elison W, Tran S, Djulamsah J, Lie A, Loe T, Holman AR, Corban S, Buchanan J, Mamde S, Zhou H, Elgamal RM, Tseliou E, Huang V, Wang Z, Chiu J, Melton R, Griffin E, Zhang Q, Lucero J, Navankasattusas S, Li D, Seng C, Destici E, Selzman CH, D’Antonio-Chronowska A, Wang T, Wang A, Drakos SG, Gaulton KJ, Ren B, Chi NC. Single cell multiomics and 3D genome architecture reveal novel pathways of human heart failure. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.05.08.25327176. [PMID: 40385400 PMCID: PMC12083629 DOI: 10.1101/2025.05.08.25327176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Heart failure is a leading cause of morbidity and mortality; yet gene regulatory mechanisms driving cell type-specific pathologic responses remain undefined. Here, we present the cell type-resolved transcriptomes, chromatin accessibility, histone modifications and chromatin organization of 36 non-failing and failing human hearts profiled from 776,479 cells spanning all cardiac chambers. Integrative analyses revealed dynamic changes in cell type composition, gene regulatory programs and chromatin organization, which expanded the annotation of cardiac cis-regulatory sequences by ten-fold and mapped cell type-specific enhancer-gene interactions. Cardiomyocytes and fibroblasts particularly exhibited complex disease-associated cellular states, gene regulatory programs and global chromatin reorganization. Mapping genetic association data onto cell type-specific regulatory programs revealed likely causal genetic contributors to heart failure. Together, these findings provide comprehensive, multimodal gene regulatory maps of the human heart in health and disease, offering a valuable framework for designing precise cell type-targeted therapies for treating heart failure.
Collapse
Affiliation(s)
- Yang Xie
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Luca Tucciarone
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Elie N. Farah
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Lei Chang
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Qian Yang
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Thirupura S. Shankar
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, Salt Lake City, UT, USA
| | - Weston Elison
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA
| | - Shaina Tran
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Jovina Djulamsah
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Audrey Lie
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Timothy Loe
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Alyssa R. Holman
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Sierra Corban
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Justin Buchanan
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Sainath Mamde
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Bioengineering Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Haowen Zhou
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Ruth M. Elgamal
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA
| | - Eleni Tseliou
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, Salt Lake City, UT, USA
- Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT, USA
| | - Vincent Huang
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, Salt Lake City, UT, USA
- Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT, USA
| | - Zhaoning Wang
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jeffery Chiu
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA
| | - Rebecca Melton
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA
| | - Emily Griffin
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Qingquan Zhang
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Jacinta Lucero
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Sutip Navankasattusas
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, Salt Lake City, UT, USA
| | - Daofeng Li
- Department of Genetics, The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Chanrung Seng
- Department of Genetics, The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Eugin Destici
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Craig H. Selzman
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, Salt Lake City, UT, USA
- Division of Cardiothoracic Surgery, University of Utah, Salt Lake City, UT, USA
| | - Agnieszka D’Antonio-Chronowska
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Ting Wang
- Department of Genetics, The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St Louis, MO, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Allen Wang
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Stavros G. Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, Salt Lake City, UT, USA
- Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT, USA
| | - Kyle J. Gaulton
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Bing Ren
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
- New York Genome Center, New York, NY, USA
- Department of Genetics and Development, Systems Biology, Biochemistry and Molecular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - Neil C. Chi
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA
| |
Collapse
|
18
|
Lee SE, Joo JH, Hwang HS, Chen SF, Evans D, Lee KY, Kim KH, Hyun J, Kim MS, Jung SH, Kim JJ, Lee JS, Torkamani A. Spatial transcriptional landscape of human heart failure. Eur Heart J 2025:ehaf272. [PMID: 40335066 DOI: 10.1093/eurheartj/ehaf272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/30/2024] [Accepted: 04/03/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND AND AIMS Heart failure (HF) remains a significant clinical challenge due to its diverse aetiologies and complex pathophysiology. The molecular alterations specific to distinct cell types and histological patterns during HF progression are still poorly characterized. This study aimed to explore cell-type- and histology-specific gene expression profiles in cardiomyopathies. METHODS Ninety tissue cores from 44 participants, encompassing various forms of cardiomyopathy and control samples with diverse histological features, were analysed using the GeoMx Whole Human Transcriptome Atlas. Data on cell types, clinical information, and histological features were integrated to examine gene expression profiles in cardiomyopathy. RESULTS The study characterized the cellular composition of ventricular myocardium and validated the GeoMx platform's efficiency in compartmentalizing specific cell types, demonstrating high accuracy for cardiomyocytes but limitations for endothelial cells and fibroblasts. Differentially expressed genes, including UCHL1 from cardiomyocytes, were associated with degeneration, while CCL14, ACKR1, and PLVAP from endothelial cells were linked to fibrosis. Multiplex immunohistochemistry and integrative analysis of prior sc/snRNA-seq data identified a PLVAP, ACKR1, and CCL14-positive pro-inflammatory endothelial cell subtype linked to fibrosis in HF. Downregulation of ribosomal proteins in cardiomyocytes was associated with myocyte disarray in hypertrophic cardiomyopathy. Additionally, pronounced inflammatory responses were observed in end-stage HF. Combined histological and clinical analysis identified CRIP3, PFKFB2, and TAX1BP3 as novel contributors to HF pathogenesis. CONCLUSIONS These findings highlight the critical role of cell-enriched and histology-specific transcriptome mapping in understanding the complex pathophysiological landscape of failing hearts, offering molecular insights and potential therapeutic targets for future interventions.
Collapse
Affiliation(s)
- Sang Eun Lee
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Scripps Research Translational Institute, 3344 North Torrey Pines Court, La Jolla, CA 92037, USA
| | - Jeong Ho Joo
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Hee Sang Hwang
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Shang-Fu Chen
- Scripps Research Translational Institute, 3344 North Torrey Pines Court, La Jolla, CA 92037, USA
| | - Douglas Evans
- Scripps Research Translational Institute, 3344 North Torrey Pines Court, La Jolla, CA 92037, USA
| | - Kyoung Yul Lee
- Pathology Center, Seegene Medical Foundation, Seoul, Korea
| | - Kyung-Hee Kim
- Division of Cardiology, Cardiovascular Center, Incheon Sejong Hospital, Incheon, Korea
| | - Junho Hyun
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Min-Seok Kim
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sung-Ho Jung
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae-Joong Kim
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jeong Seok Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Ali Torkamani
- Scripps Research Translational Institute, 3344 North Torrey Pines Court, La Jolla, CA 92037, USA
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| |
Collapse
|
19
|
Xu S, Li H, Han J, Xu Y, Li N, Che W, Liu F, Yue W. Klf9 promotes the repair of myocardial infarction by regulating macrophage recruitment and polarization. JCI Insight 2025; 10:e187072. [PMID: 40198141 DOI: 10.1172/jci.insight.187072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
The inflammatory response after myocardial infarction (MI) is a precisely regulated process that greatly affects subsequent wound healing and remodeling. However, understanding about the process is still limited. Macrophages are critically involved in inflammation resolution after MI. Krüppel-like factor 9 (Klf9) is a C2H2 zinc finger-containing transcription factor that has been implicated in glucocorticoid regulation of macrophages. However, the contribution of Klf9 to macrophage phenotype and function in the context of MI remains unclear. Our study revealed that KLF9 deficiency resulted in higher mortality and cardiac rupture rate, as well as a considerable exacerbation in cardiac function. Single-cell RNA sequencing and flow cytometry analyses revealed that, compared with WT mice, Klf9-/- mice displayed excessive neutrophil infiltration, insufficient macrophage infiltration, and a reduced proportion of monocyte-derived CD206+ macrophages after MI. Moreover, the expression of IFN-γ/STAT1 pathway genes in Klf9-/- cardiac macrophages was dysregulated, characterized by insufficient expression at 1 day post-MI and excessive expression at day 3 post-MI. Mechanistically, Klf9 directly binds to the promoters of Stat1 gene, regulating its transcription. Overall, these findings indicate that Klf9 beneficially influences wound healing after MI by modulating macrophage recruitment and differentiation by regulating the IFN-γ/STAT1 signaling pathway.
Collapse
Affiliation(s)
- Sheng Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hao Li
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jun Han
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Niannian Li
- Department of Otolaryngology Head and Neck Surgery, Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, China
| | - Wenliang Che
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Feng Liu
- Department of Otolaryngology Head and Neck Surgery, Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, China
| | - Wenhui Yue
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Zhou Y, Sheng Q, Jin S. Integrating single-cell data with biological variables. Proc Natl Acad Sci U S A 2025; 122:e2416516122. [PMID: 40294274 PMCID: PMC12067276 DOI: 10.1073/pnas.2416516122] [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: 08/15/2024] [Accepted: 03/30/2025] [Indexed: 04/30/2025] Open
Abstract
Constructing single-cell atlases requires preserving differences attributable to biological variables, such as cell types, tissue origins, and disease states, while eliminating batch effects. However, existing methods are inadequate in explicitly modeling these biological variables. Here, we introduce SIGNAL, a general framework that leverages biological variables to disentangle biological and technical effects, thereby linking these metadata to data integration. SIGNAL employs a variant of principal component analysis to align multiple batches, enabling the integration of 1 million cells in approximately 2 min. SIGNAL, despite its computational simplicity, surpasses state-of-the-art methods across multiple integration scenarios: 1) heterogeneous datasets, 2) cross-species datasets, 3) simulated datasets, 4) integration on low-quality cell annotations, and 5) reference-based integration. Furthermore, we demonstrate that SIGNAL accurately transfers knowledge from reference to query datasets. Notably, we propose a self-adjustment strategy to restore annotated cell labels potentially distorted during integration. Finally, we apply SIGNAL to multiple large-scale atlases, including a human heart cell atlas containing 2.7 million cells, identifying tissue- and developmental stage-specific subtypes, as well as condition-specific cell states. This underscores SIGNAL's exceptional capability in multiscale analysis.
Collapse
Affiliation(s)
- Yang Zhou
- School of Mathematics, Harbin Institute of Technology, Harbin150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou450000, China
| | - Qiongyu Sheng
- School of Mathematics, Harbin Institute of Technology, Harbin150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou450000, China
| | - Shuilin Jin
- School of Mathematics, Harbin Institute of Technology, Harbin150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou450000, China
| |
Collapse
|
21
|
Nagai JS, Maié T, Schaub MT, Costa IG. scACCorDiON: a clustering approach for explainable patient level cell-cell communication graph analysis. Bioinformatics 2025; 41:btaf288. [PMID: 40327499 PMCID: PMC12122081 DOI: 10.1093/bioinformatics/btaf288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/08/2025] Open
Abstract
MOTIVATION Combining single-cell sequencing with ligand-receptor (LR) analysis paves the way for the characterization of cell communication events in complex tissues. In particular, directed weighted graphs naturally represent cell-cell communication events. However, current computational methods cannot yet analyze sample-specific cell-cell communication events, as measured in single-cell data produced in large patient cohorts. Cohort-based cell-cell communication analysis presents many challenges, such as the nonlinear nature of cell-cell communication and the high variability given by the patient-specific single-cell RNAseq datasets. RESULTS Here, we present scACCorDiON (single-cell Analysis of Cell-Cell Communication in Disease clusters using Optimal transport in Directed Networks), an optimal transport algorithm exploring node distances on the Markov Chain as the ground metric between directed weighted graphs. Benchmarking indicates that scACCorDiON performs a better clustering of samples according to their disease status than competing methods that use undirected graphs. We provide a case study of pancreas adenocarcinoma, where scACCorDion detects a sub-cluster of disease samples associated with changes in the tumor microenvironment. Our study case corroborates that clusters provide a robust and explainable representation of cell-cell communication events and that the expression of detected LR pairs is predictive of pancreatic cancer survival. AVAILABILITY AND IMPLEMENTATION The code of scACCorDiON is available at https://scaccordion.readthedocs.io/en/latest/. and https://doi.org/10.5281/zenodo.15267648. The survival analysis package can be found at https://github.com/CostaLab/scACCorDiON.su.
Collapse
Affiliation(s)
- James S Nagai
- Institute for Computational Genomics, RWTH Aachen Medical Faculty, Aachen 52074, Germany
| | - Tiago Maié
- Institute for Computational Genomics, RWTH Aachen Medical Faculty, Aachen 52074, Germany
| | - Michael T Schaub
- Department of Computational Science, RWTH Aachen University, Aachen 52074, Germany
| | - Ivan G Costa
- Institute for Computational Genomics, RWTH Aachen Medical Faculty, Aachen 52074, Germany
| |
Collapse
|
22
|
Sun Y, Xu H, Zhu Y, Rao Y, Fan X, Wang Z, Gu H, Yue X, Zhao X, Su L, Cai R. Single-cell and spatial transcriptomic analyses reveal transcriptional cell lineage heterogeneity in extracranial arteriovenous malformation. J Dermatol Sci 2025; 118:66-75. [PMID: 40118698 DOI: 10.1016/j.jdermsci.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 02/05/2025] [Accepted: 02/20/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Extracranial arteriovenous malformations (eAVMs) are rare congenital vascular anomalies consisting of abnormal artery-vein bypass with no intervening capillary network, and can lead to disability and death. The critical genetic determination factors and key transcriptional pathways of the eAVMs genesis process are still unclear. OBJECTIVE To generate an overview of the molecular information within eAVMs at the single-cell level. METHODS We performed single-cell RNA sequencing (scRNA-seq) on nine samples of eAVMs receiving a confirmatory histopathologic evaluation from a board-certified dermatopathologist and two nonlesional tissue sample controls. 10x Visium spatial transcriptomics (ST) was performed on one eAVM to spatially localize heterogeneous cells and profile the gene expression dynamics of the cells in their morphological context. The scRNA-seq and ST data were integrated and analyzed to further query for spatially restricted mapping of intrapopulation heterogeneous cells. RESULTS We identified different cell states of endothelial cells (ECs), perivascular cells and immune cells in eAVMs, uncovered the presence of MAFB+ nidus ECs, characterized mesenchymal activation in ECs, and identified transcriptional variation within perivascular cells and the presence of smooth muscle-like pericytes in eAVMs. Dysregulated cell to cell interactions among ECs, perivascular cells and immune cells that are associated with eAVMs, including those involving MDK, VEGF, ANGPT, SEMA3 and GALECTIN-9 were cataloged. Together, our results depicted the heterogeneity underlying cell function and interaction of eAVMs at a single-cell resolution. CONCLUSION We present a comprehensive picture of the cell-resolution atlas that describes the transcriptomic heterogeneity underlying cell function and interaction in eAVMs.
Collapse
Affiliation(s)
- Yi Sun
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoyang Xu
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanze Zhu
- School of Automation, Northwestern Polytechnical University, Xi 'an, China
| | - Yamin Rao
- Department of Pathology, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xindong Fan
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenfeng Wang
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Gu
- Department of Burn and Plastic Surgery, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiaojie Yue
- Department of Burn and Plastic Surgery, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiong Zhao
- Department of Burn and Plastic Surgery, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Lixin Su
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ren Cai
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
23
|
Biondi-Zoccai G, Weber BN, Abbate A, Booz GW. Cardiac Immunotherapy, Immuno-Cardiology, and the Future of Cardiovascular Pharmacology. J Cardiovasc Pharmacol 2025; 85:308-311. [PMID: 40014789 DOI: 10.1097/fjc.0000000000001687] [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] [Received: 12/18/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
ABSTRACT Immuno-cardiology is an emerging field that explores the interplay between the immune system, inflammation, and cardiovascular health/disease, aiming to develop innovative therapies for preventing and treating cardiac diseases. Indeed, chronic inflammation and immune dysregulation play pivotal roles in most cardiovascular conditions, including arrhythmias, atherothrombosis, ischemic heart disease, heart failure, and valve disease. Recent advances in immune-based therapies, including chimeric antigen receptor-T and chimeric antigen receptor-macrophage technologies, have demonstrated potential in impacting on cardiac fibrosis and thus improving surrogate end points in preclinical studies. Immune checkpoint inhibitors, while already established as an effective intervention in oncology, present challenges in their cardiovascular applications because of cardiotoxic side effects, highlighting the need for dedicated cardioprotective strategies or further molecular refinements. Nanoparticle-based delivery systems and cytokine-targeted therapies may offer precise modulation of immune responses, while gut microbiota interventions could exploit the systemic impact of inflammation on cardiovascular health. Despite these quite promising advances, barriers such as safety, scalability, and patient-specific responses must be addressed, and thus precision and personalized approaches will be crucial to overcoming these challenges and ensuring safe and also equitable access. By leveraging interdisciplinary collaboration and technological innovations, immuno-cardiology holds the promise of transforming the prevention and treatment landscape for cardiac diseases, paving the way for improved outcomes and quality of life for patients worldwide.
Collapse
Affiliation(s)
- Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Brittany N Weber
- Heart and Vascular Center, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Antonio Abbate
- Berne Cardiovascular Research Center and Division of Cardiology, University of Virginia, Charlottesville, VA; and
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
24
|
Jogani S, Pol AS, Prajapati M, Samal A, Bhatia K, Parmar J, Patel U, Shah F, Vyas N, Gupta S. scaLR: a low-resource deep neural network-based platform for single cell analysis and biomarker discovery. Brief Bioinform 2025; 26:bbaf243. [PMID: 40439670 PMCID: PMC12121358 DOI: 10.1093/bib/bbaf243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/14/2025] [Accepted: 05/02/2025] [Indexed: 06/02/2025] Open
Abstract
Single-cell ribonucleic acid (RNA) sequencing (scRNA-seq) produces vast amounts of individual cell profiling data. Its analysis presents a significant challenge in accurately annotating cell types and their associated biomarkers. Different pipelines based on deep neural network (DNN) methods have been employed to tackle these issues. These pipelines have arisen as a promising resource and can extract meaningful and concise features from noisy, diverse, and high-dimensional data to enhance annotations and subsequent analysis. Existing tools require high computational resources to execute large sample datasets. We have developed a cutting-edge platform known as scaLR (Single-cell analysis using low resource) that efficiently processes data into feature subsets, samples in batches to reduce the required memory for processing large datasets, and running DNN models in multiple central processing units. scaLR is equipped with data processing, feature extraction, training, evaluation, and downstream analysis. Its novel feature extraction algorithm first trains the model on a feature subset and stores the importance of the features for all the features in that subset. At the end of the training of all subsets, the top-K features are selected based on their importance. The final model is trained on top-K features; its performance evaluation and associated downstream analysis provide significant biomarkers for different cell types and diseases/traits. Our findings indicate that scaLR offers comparable prediction accuracy and requires less model training time and computational resources than existing Python-based pipelines. We present scaLR, a Python-based platform, engineered to utilize minimal computational resources while maintaining comparable execution times and analysis costs to existing frameworks.
Collapse
Affiliation(s)
- Saiyam Jogani
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Laxman Nagar Baner, Pune 411045, Maharashtra, India
| | - Anand Santosh Pol
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Laxman Nagar Baner, Pune 411045, Maharashtra, India
| | - Mayur Prajapati
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| | - Amit Samal
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| | - Kriti Bhatia
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Laxman Nagar Baner, Pune 411045, Maharashtra, India
| | - Jayendra Parmar
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| | - Urvik Patel
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| | - Falak Shah
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| | - Nisarg Vyas
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| | - Saurabh Gupta
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| |
Collapse
|
25
|
Torregrossa M, Davies L, Hans-Günther M, Simon JC, Franz S, Rinkevich Y. Effects of embryonic origin, tissue cues and pathological signals on fibroblast diversity in humans. Nat Cell Biol 2025; 27:720-735. [PMID: 40263573 DOI: 10.1038/s41556-025-01638-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/18/2025] [Indexed: 04/24/2025]
Abstract
Fibroblasts, once perceived as a uniform cell type, are now recognized as a mosaic of distinct populations with specialized roles in tissue homeostasis and pathology. Here we provide a global overview of the expanding compendium of fibroblast cell types and states, their diverse lineage origins and multifaceted functions across various human organs. By integrating insights from developmental biology, lineage tracing and single-cell technologies, we highlight the complex nature of fibroblasts. We delve into their origination from embryonic mesenchyme and tissue-resident populations, elucidating lineage-specific behaviours in response to physiological cues. Furthermore, we highlight the pivotal role of fibroblasts in orchestrating tissue repair, connective tissue remodelling and immune modulation across diverse pathologies. This knowledge is essential to develop novel fibroblast-targeted therapies to restore steady-state fibroblast function and advance regenerative medicine strategies across multiple diseases.
Collapse
Affiliation(s)
- Marta Torregrossa
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Faculty, Leipzig, Germany
| | - Lindsay Davies
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Machens Hans-Günther
- Department for Plastic Surgery and Hand Surgery, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Jan C Simon
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Faculty, Leipzig, Germany
| | - Sandra Franz
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Faculty, Leipzig, Germany.
| | - Yuval Rinkevich
- Chinese Institutes for Medical Research, Beijing, China.
- Capital Medical University, Beijing, China.
| |
Collapse
|
26
|
Fan Y, Zheng Y, Zhang Y, Xu G, Liu C, Hu J, Ji Q, Zhang S, Fang S, Lei J, Li LZ, Wang X, Xu X, Wang C, Wang S, Ma S, Song M, Jiang W, Zhu J, Feng Y, Wang J, Yang Y, Zhu G, Tian XL, Zhang H, Song W, Yang J, Yao Y, Liu GH, Qu J, Zhang W. ARID5A orchestrates cardiac aging and inflammation through MAVS mRNA stabilization. NATURE CARDIOVASCULAR RESEARCH 2025; 4:602-623. [PMID: 40301689 DOI: 10.1038/s44161-025-00635-z] [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: 09/08/2023] [Accepted: 03/10/2025] [Indexed: 05/01/2025]
Abstract
Elucidating the regulatory mechanisms of human cardiac aging remains a great challenge. Here, using human heart tissues from 74 individuals ranging from young (≤35 years) to old (≥65 years), we provide an overview of the histological, cellular and molecular alterations underpinning the aging of human hearts. We decoded aging-related gene expression changes at single-cell resolution and identified increased inflammation as the key event, driven by upregulation of ARID5A, an RNA-binding protein. ARID5A epi-transcriptionally regulated Mitochondrial Antiviral Signaling Protein (MAVS) mRNA stability, leading to NF-κB and TBK1 activation, amplifying aging and inflammation phenotypes. The application of gene therapy using lentiviral vectors encoding shRNA targeting ARID5A into the myocardium not only mitigated the inflammatory and aging phenotypes but also bolstered cardiac function in aged mice. Altogether, our study provides a valuable resource and advances our understanding of cardiac aging mechanisms by deciphering the ARID5A-MAVS axis in post-transcriptional regulation.
Collapse
Affiliation(s)
- Yanling Fan
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Yandong Zheng
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yiyuan Zhang
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Gang Xu
- Liver Transplant Center, Organ Transplant Center, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital of Sichuan University, Chengdu, China
| | - Chun Liu
- Department of Physiology and Medicine, Cardiovascular Center, Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jianli Hu
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qianzhao Ji
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuo Zhang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuaiqi Fang
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jinghui Lei
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Lan-Zhu Li
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xing Wang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xi Xu
- Liver Transplant Center, Organ Transplant Center, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital of Sichuan University, Chengdu, China
| | - Cui Wang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Si Wang
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shuai Ma
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Aging Biomarker Consortium, Beijing, China
| | - Moshi Song
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Wenjian Jiang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Junming Zhu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yijia Feng
- Oujiang Laboratory, Center for Geriatric Medicine and Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research for Mental Disorders, The First-affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiangang Wang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ying Yang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Guodong Zhu
- Institute of Gerontology, Guangzhou Geriatric Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Li Tian
- Aging and Vascular Diseases, Human Aging Research Institute and School of Life Science, Nanchang University and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Hongjia Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Weihong Song
- Oujiang Laboratory, Center for Geriatric Medicine and Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research for Mental Disorders, The First-affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiayin Yang
- Liver Transplant Center, Organ Transplant Center, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital of Sichuan University, Chengdu, China
| | - Yan Yao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Guang-Hui Liu
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| | - Jing Qu
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
- Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Weiqi Zhang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| |
Collapse
|
27
|
Guo CK, Xia CR, Peng G, Cao ZJ, Gao G. Learning Phenotype Associated Signature in Spatial Transcriptomics with PASSAGE. SMALL METHODS 2025; 9:e2401451. [PMID: 39905872 DOI: 10.1002/smtd.202401451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/31/2024] [Indexed: 02/06/2025]
Abstract
Spatially resolved transcriptomics (SRT) is poised to advance the understanding of cellular organization within complex tissues under various physiological and pathological conditions at unprecedented resolution. Despite the development of numerous computational tools that facilitate the automatic identification of statistically significant intra-/inter-slice patterns (like spatial domains), these methods typically operate in an unsupervised manner, without leveraging sample characteristics like physiological/pathological states. Here PASSAGE (Phenotype Associated Spatial Signature Analysis with Graph-based Embedding), a rationally-designed deep learning framework is presented for characterizing phenotype-associated signatures across multiple heterogeneous spatial slices effectively. In addition to its outstanding performance in systematic benchmarks, PASSAGE's unique capability in calling sophisticated signatures has been demonstrated in multiple real-world cases. The full package of PASSAGE is available at https://github.com/gao-lab/PASSAGE.
Collapse
Affiliation(s)
- Chen-Kai Guo
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, University of Chinese Academy of Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chen-Rui Xia
- State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Biomedical Pioneering Innovative Center (BIOPIC) and Beijing Advanced Innovation Center for Genomics (ICG), Center for Bioinformatics (CBI), Peking University, Beijing, 100871, China
- Changping Laboratory, Beijing, 102206, China
| | - Guangdun Peng
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, University of Chinese Academy of Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhi-Jie Cao
- State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Biomedical Pioneering Innovative Center (BIOPIC) and Beijing Advanced Innovation Center for Genomics (ICG), Center for Bioinformatics (CBI), Peking University, Beijing, 100871, China
- Changping Laboratory, Beijing, 102206, China
| | - Ge Gao
- State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Biomedical Pioneering Innovative Center (BIOPIC) and Beijing Advanced Innovation Center for Genomics (ICG), Center for Bioinformatics (CBI), Peking University, Beijing, 100871, China
- Changping Laboratory, Beijing, 102206, China
| |
Collapse
|
28
|
Byatt TC, Razaghi E, Tüzüner S, Simões FC. Immune-mediated cardiac development and regeneration. Semin Cell Dev Biol 2025; 171:103613. [PMID: 40315634 DOI: 10.1016/j.semcdb.2025.103613] [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: 12/29/2024] [Revised: 03/18/2025] [Accepted: 04/16/2025] [Indexed: 05/04/2025]
Abstract
The complex interplay between the immune and cardiovascular systems during development, homeostasis and regeneration represents a rapidly evolving field in cardiac biology. Single cell technologies, spatial mapping and computational analysis have revolutionised our understanding of the diversity and functional specialisation of immune cells within the heart. From the earliest stages of cardiogenesis, where primitive macrophages guide heart tube formation, to the complex choreography of inflammation and its resolution during regeneration, immune cells emerge as central orchestrators of cardiac fate. Translating these fundamental insights into clinical applications represents a major challenge and opportunity for the field. In this Review, we decode the immunological blueprint of heart development and regeneration to transform cardiovascular disease treatment and unlock the regenerative capacity of the human heart.
Collapse
Affiliation(s)
- Timothy C Byatt
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Ehsan Razaghi
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Selin Tüzüner
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Filipa C Simões
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
29
|
Yang S, Yan L, Chen L, Su G, Yang L, Gong L, Liu L. Cardiac PDK4 promotes neutrophilic PFKL methylation and drives the innate immune response in diabetic myocardial infarction. Pharmacol Res 2025; 215:107731. [PMID: 40222696 DOI: 10.1016/j.phrs.2025.107731] [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] [Received: 12/13/2024] [Revised: 03/20/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
NETosis plays a pivotal role in the innate immune response after diabetic myocardial infarction (MI), exerting a profound influence on the overall pathological process and potential recovery outcomes. The metabolism of diabetic cardiomyocyte actively creates a specialized micro environment for the innate immune response after MI. However, the mechanism by which cardiac metabolism drives NETosis remains unclear. Utilizing public databases of human MI sc-RNA datasets, we discovered that cardiomyocyte PDK4 expression mediates the intensification of glycolysis, which is strongly correlated with NETosis. Through mass spectrometry imaging and phenotype assessment, we ascertained that specific knockout of PDK4 in cardiomyocytes (PDK4fl/flMyh6Cre, male, 6 weeks) led to a reduction in NETosis by restraining micro environmental lactate (LA) production. In addition, the role of LA in promoting NETosis has been further corroborated by in vivo/in vitro experiments involving LA supplementation and its absence. Moreover, LA redirects neutrophil metabolic flux from glycolysis to the pentose-phosphate pathway (PPP). Mechanistically, LA triggers metabolic remodeling through the PRMT9-mediated methylation of PFKL at the R301 residue, resulting in PFKL inactivation and the consequent restriction of glycolysis. Our findings reveal the crucial role of cardiomyocyte metabolism in NETosis, shedding light on the role of LA as a vital signaling molecule in the crosstalk between cardiomyocytes and neutrophils. Importantly, we screened pitavastatin, a potential inhibitor of PDK4 among the FDA-approved drugs, and verified that it can alleviate NETosis in diabetic MI, which provides a rationale for drug selection in diabetic MI patients.
Collapse
Affiliation(s)
- Song Yang
- China-Japan Friendship Hospital, Beijing 100029, China
| | - Longxin Yan
- China-Japan Friendship Hospital, Beijing 100029, China
| | - Lang Chen
- China-Japan Friendship Hospital, Beijing 100029, China
| | - Gaijuan Su
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100007, China
| | - Long Yang
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Lili Gong
- China-Japan Friendship Hospital, Beijing 100029, China.
| | - Lihong Liu
- China-Japan Friendship Hospital, Beijing 100029, China.
| |
Collapse
|
30
|
Pekayvaz K, Heinig M, Stark K. Predictive cardio-omics: translating single-cell multiomics into tools for personalized medicine. Nat Rev Cardiol 2025; 22:305-306. [PMID: 39900732 DOI: 10.1038/s41569-025-01132-3] [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: 02/05/2025]
Affiliation(s)
- Kami Pekayvaz
- Medizinische Klinik und Poliklinik I, LMU University Hospital, Munich, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
| | - Matthias Heinig
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
- Institute of Computational Biology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany.
- Department of Computer Science, TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany.
| | - Konstantin Stark
- Medizinische Klinik und Poliklinik I, LMU University Hospital, Munich, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
31
|
Tanevski J, Vulliard L, Ibarra-Arellano MA, Schapiro D, Hartmann FJ, Saez-Rodriguez J. Learning tissue representation by identification of persistent local patterns in spatial omics data. Nat Commun 2025; 16:4071. [PMID: 40307222 PMCID: PMC12044154 DOI: 10.1038/s41467-025-59448-0] [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: 03/26/2024] [Accepted: 04/23/2025] [Indexed: 05/02/2025] Open
Abstract
Spatial omics data provide rich molecular and structural information on tissues. Their analysis provides insights into local heterogeneity of tissues and holds promise to improve patient stratification by associating clinical observations with refined tissue representations. We introduce Kasumi, a method for identifying spatially localized neighborhood patterns of intra- and intercellular relationships that are persistent across samples and conditions. The tissue representation based on these patterns can facilitate translational tasks, as we show for stratification of cancer patients for disease progression and response to treatment using data from different experimental platforms. On these tasks, Kasumi outperforms related approaches and offers explanations of spatial coordination and relationships at the cell-type or marker level. We show that persistent patterns comprise regions of different sizes, and that non-abundant, localized relationships in the tissue are strongly associated with unfavorable outcomes.
Collapse
Affiliation(s)
- Jovan Tanevski
- Institute for Computational Biomedicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany.
- Translational Spatial Profiling Center, Heidelberg University Hospital, Heidelberg, Germany.
- Department of Knowledge Technologies, Jožef Stefan Institute, Ljubljana, Slovenia.
| | - Loan Vulliard
- Institute for Computational Biomedicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
- Systems Immunology and Single-Cell Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Miguel A Ibarra-Arellano
- Institute for Computational Biomedicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
| | - Denis Schapiro
- Institute for Computational Biomedicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
- Translational Spatial Profiling Center, Heidelberg University Hospital, Heidelberg, Germany
- Institute of Pathology, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
| | - Felix J Hartmann
- Systems Immunology and Single-Cell Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Julio Saez-Rodriguez
- Institute for Computational Biomedicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany.
- Translational Spatial Profiling Center, Heidelberg University Hospital, Heidelberg, Germany.
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK.
| |
Collapse
|
32
|
Bahr J, Poschmann G, Jungmann A, Busch M, Ding Z, Vogt J, Zalfen R, Steinhausen J, Euan Martínez AA, Wachtmeister T, Rickert D, Lautwein T, Alter C, Amrute JM, Lavine KJ, Köhrer K, Levkau B, Most P, Stühler K, Hesse J, Schrader J. A secretome atlas of cardiac fibroblasts from healthy and infarcted mouse hearts. Commun Biol 2025; 8:675. [PMID: 40301568 PMCID: PMC12041564 DOI: 10.1038/s42003-025-08083-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: 07/31/2024] [Accepted: 04/12/2025] [Indexed: 05/01/2025] Open
Abstract
Cardiac fibroblasts (CF) are key players after myocardial infarction (MI), but their signaling is only incompletely understood. Here we report a first secretome atlas of CF in control (cCF) and post-MI mouse hearts (miCF), combining a rapid cell isolation technique with SILAC and click chemistry. In CF, numerous paracrine factors involved in immune homeostasis are identified. Comparing secretome, transcriptome (SLAMseq), and cellular proteome disclose protein turnover. In miCF at day 5 post-MI, significantly upregulated proteins include SLIT2, FN1, and CRLF1 in mouse and human samples. Comparing the miCF secretome at days 3 and 5 post-MI reveals the dynamic nature of protein secretion. Specific in-vivo labeling of miCF proteins via biotin ligase TurboID using the POSTN promotor mirrors the in-vitro data. In summary, we identify numerous paracrine factors specifically secreted from CF in mice and humans. This secretome atlas may lead to new biomarkers and/or therapeutic targets for the activated CF.
Collapse
Affiliation(s)
- Jasmin Bahr
- Department of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Gereon Poschmann
- Institute for Molecular Medicine, Proteome Research, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andreas Jungmann
- Division of Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Busch
- Division of Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
| | - Zhaoping Ding
- Department of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jens Vogt
- Institute of Molecular Medicine III, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ria Zalfen
- Department of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Julia Steinhausen
- Department of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Arlen Aurora Euan Martínez
- Department of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Thorsten Wachtmeister
- Genomics & Transcriptomics Laboratory, Biological and Medical Research Centre (BMFZ), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Daniel Rickert
- Genomics & Transcriptomics Laboratory, Biological and Medical Research Centre (BMFZ), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tobias Lautwein
- Genomics & Transcriptomics Laboratory, Biological and Medical Research Centre (BMFZ), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christina Alter
- Department of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Junedh M Amrute
- Center for Cardiovascular Research, Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Kory J Lavine
- Center for Cardiovascular Research, Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Karl Köhrer
- Genomics & Transcriptomics Laboratory, Biological and Medical Research Centre (BMFZ), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Bodo Levkau
- Institute of Molecular Medicine III, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Patrick Most
- Division of Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
| | - Kai Stühler
- Institute for Molecular Medicine, Proteome Research, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Molecular Proteomics Laboratory, Biological and Medical Research Centre (BMFZ), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Julia Hesse
- Department of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| | - Jürgen Schrader
- Department of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
33
|
Li L, Lu M, Guo L, Zhang X, Liu Q, Zhang M, Gao J, Xu M, Lu Y, Zhang F, Li Y, Zhang R, Liu X, Pan S, Zhang X, Li Z, Chen Y, Su X, Zhang N, Guo W, Yang T, Chen J, Qin Y, Zhang Z, Cui W, Yu L, Gu Y, Yang H, Xu X, Wang J, Burns CE, Burns CG, Han K, Zhao L, Fan G, Su Y. An organ-wide spatiotemporal transcriptomic and cellular atlas of the regenerating zebrafish heart. Nat Commun 2025; 16:3716. [PMID: 40253397 PMCID: PMC12009352 DOI: 10.1038/s41467-025-59070-0] [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: 06/12/2024] [Accepted: 04/10/2025] [Indexed: 04/21/2025] Open
Abstract
Adult zebrafish robustly regenerate injured hearts through a complex orchestration of molecular and cellular activities. However, this remarkable process, which is largely non-existent in humans, remains incompletely understood. Here, we utilize integrated spatial transcriptomics (Stereo-seq) and single-cell RNA-sequencing (scRNA-seq) to generate a spatially-resolved molecular and cellular atlas of regenerating zebrafish heart across eight stages. We characterize the cascade of cardiomyocyte cell states responsible for producing regenerated myocardium and explore a potential role for tpm4a in cardiomyocyte re-differentiation. Moreover, we uncover the activation of ifrd1 and atp6ap2 genes as a unique feature of regenerative hearts. Lastly, we reconstruct a 4D "virtual regenerating heart" comprising 569,896 cells/spots derived from 36 scRNA-seq libraries and 224 Stereo-seq slices. Our comprehensive atlas serves as a valuable resource to the cardiovascular and regeneration scientific communities and their ongoing efforts to understand the molecular and cellular mechanisms underlying vertebrate heart regeneration.
Collapse
Affiliation(s)
- Lei Li
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
| | - Meina Lu
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Lidong Guo
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuejiao Zhang
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Qun Liu
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Meiling Zhang
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Junying Gao
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Mengyang Xu
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
| | - Yijian Lu
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Fang Zhang
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Yao Li
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Ruihua Zhang
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Xiawei Liu
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Shanshan Pan
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Xianghui Zhang
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Zhen Li
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Yadong Chen
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Xiaoshan Su
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Nannan Zhang
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Wenjie Guo
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Tao Yang
- China National GeneBank, BGI Research, Shenzhen, 518120, China
| | - Jing Chen
- China National GeneBank, BGI Research, Shenzhen, 518120, China
| | - Yating Qin
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | | | - Wei Cui
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Lindong Yu
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Ying Gu
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Huanming Yang
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Xun Xu
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Jianxun Wang
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Caroline E Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - C Geoffrey Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Kai Han
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China.
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark.
| | - Long Zhao
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China.
- College of Fisheries, Ocean University of China, Qingdao, 266003, China.
| | - Guangyi Fan
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China.
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China.
- BGI Research, Sanya, 572025, China.
- BGI Research, Hangzhou, 310030, China.
| | - Ying Su
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China.
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
34
|
He X, Wang Q, Long Q, Zhong Y, Qi Z, Zhang Y, Chang L, Qian B, Huang S, Wang X, Chen X, Li F, Yang X, Gao WD, Song Z, Xu L, Zhao Q. The single-cell atlas of short-chain fatty acid receptors in human and mice hearts. Front Immunol 2025; 16:1538384. [PMID: 40308581 PMCID: PMC12040890 DOI: 10.3389/fimmu.2025.1538384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/18/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction The gut microbiota metabolite, short-chain fatty acids (SCFAs), can protect against multiple cardiovascular diseases, while the molecular targets and underlying mechanisms need to be elucidated. One of the primary mechanisms of SCFA benefits was the direct activation of a group of G-protein-coupled receptors (GPCRs), termed free fatty acid receptors (FFARs), the FFAR2 (GPR43), and FFAR3 (GPR41). At present, the distribution of FFAR2/3 in cardiac cells has not been entirely clarified. Methods Using 18 public single-cell RNA-seq and single-nuclear RNA-seq data of human and mouse hearts, we illustrate the entire atlas of FFAR2/3 distribution in different regions and cell types in normal and infarcted hearts. Results and discussion We present the atlas of FFAR2/3 in the whole human body, normal and infarcted hearts at single-cell resolution. We also illustrated the entire atlas of FFAR2/3 in normal/ischemic hearts of newborn and adult mice by combining public and newly built sc/snRNA-seq datasets. These findings provide valuable information on the possible effect of SCFAs via FFAR2/3 in the heart and valuable references for future studies.
Collapse
Affiliation(s)
- Xiaojun He
- Cardiovascular Medical Center, Department of Cardiovascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiang Wang
- Cardiovascular Medical Center, Department of Cardiovascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qiang Long
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yiming Zhong
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhaoxi Qi
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yecen Zhang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lan Chang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bei Qian
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shixing Huang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xinming Wang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xuemei Chen
- Department of Anesthesiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Feifei Li
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaomei Yang
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Cardiology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Wei Dong Gao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Zhizhao Song
- Cardiovascular Medical Center, Department of Cardiovascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Li Xu
- Clinical Trial Institution, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Qiang Zhao
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
35
|
Senesi G, Lodrini AM, Mohammed S, Mosole S, Hjortnaes J, Veltrop RJA, Kubat B, Ceresa D, Bolis S, Raimondi A, Torre T, Malatesta P, Goumans MJ, Paneni F, Camici GG, Barile L, Balbi C, Vassalli G. miR-24-3p secreted as extracellular vesicle cargo by cardiomyocytes inhibits fibrosis in human cardiac microtissues. Cardiovasc Res 2025; 121:143-156. [PMID: 39527589 PMCID: PMC11998913 DOI: 10.1093/cvr/cvae243] [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: 04/17/2024] [Revised: 08/19/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
AIMS Cardiac fibrosis in response to injury leads to myocardial stiffness and heart failure. At the cellular level, fibrosis is triggered by the conversion of cardiac fibroblasts (CF) into extracellular matrix-producing myofibroblasts. miR-24-3p regulates this process in animal models. Here, we investigated whether miR-24-3p plays similar roles in human models. METHODS AND RESULTS Gain- and loss-of-function experiments were performed using human induced pluripotent stem cell-derived cardiomyocytes (hCM) and primary hCF under normoxic or ischaemia-simulating conditions. hCM-derived extracellular vesicles (EVs) were added to hCF. Similar experiments were performed using three-dimensional human cardiac microtissues and ex vivo cultured human cardiac slices. hCF transfection with miR-24-3p mimic prevented TGFβ1-mediated induction of FURIN, CCND1, and SMAD4-miR-24-3p target genes participating in TGFβ1-dependent fibrogenesis-regulating hCF-to-myofibroblast conversion. hCM secreted miR-24-3p as EV cargo. hCM-derived EVs modulated hCF activation. Ischaemia-simulating conditions induced miR-24-3p depletion in hCM-EVs and microtissues. Similarly, hypoxia down-regulated miR-24-3p in cardiac slices. Analyses of clinical samples revealed decreased miR-24-3p levels in circulating EVs in patients with acute myocardial infarction (AMI), compared with healthy subjects. Post-mortem RNAScope analysis showed miR-24-3p down-regulation in myocardium from patients with AMI, compared with patients who died from non-cardiac diseases. Berberine, a plant-derived agent with miR-24-3p-stimulatory activity, increased miR-24-3p contents in hCM-EVs, down-regulated FURIN, CCND1, and SMAD4, and inhibited fibrosis in cardiac microtissues. CONCLUSION These findings suggest that hCM may control hCF activation through miR-24-3p secreted as EV cargo. Ischaemia impairs this mechanism, favouring fibrosis.
Collapse
Affiliation(s)
- Giorgia Senesi
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Istituto Cardiocentro Ticino, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
| | - Alessandra M Lodrini
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Shafeeq Mohammed
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Zurich, Switzerland
| | - Simone Mosole
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland (IOSI), Switzerland
| | - Jesper Hjortnaes
- Department of Thoracic Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Rogier J A Veltrop
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
| | - Bela Kubat
- Department of Pathology, Maastricht University Medical Center, The Netherlands
| | - Davide Ceresa
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Sara Bolis
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Istituto Cardiocentro Ticino, Bellinzona, Switzerland
| | - Andrea Raimondi
- Institute of Biomedical Research, IRB, Bellinzona, Switzerland
| | - Tiziano Torre
- Heart Surgery Unit, Cardiocentro Ticino Institute, EOC, Lugano, Switzerland
| | - Paolo Malatesta
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Experimental Medicine (DIMES), Experimental Biology Unit, University of Genova, Genova, Italy
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Zurich, Switzerland
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Lucio Barile
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Istituto Cardiocentro Ticino, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Faculty of Biomedical Sciences, Euler Institute, Università della Svizzera italiana, Lugano, Switzerland
| | - Carolina Balbi
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Medicine, Baden Cantonal Hospital, Baden, Switzerland
| | - Giuseppe Vassalli
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Istituto Cardiocentro Ticino, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| |
Collapse
|
36
|
Velu PP, Abhari RE, Henderson NC. Spatial genomics: Mapping the landscape of fibrosis. Sci Transl Med 2025; 17:eadm6783. [PMID: 40203082 DOI: 10.1126/scitranslmed.adm6783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 03/19/2025] [Indexed: 04/11/2025]
Abstract
Organ fibrosis causes major morbidity and mortality worldwide. Treatments for fibrosis are limited, with organ transplantation being the only cure. Here, we review how various state-of-the-art spatial genomics approaches are being deployed to interrogate fibrosis across multiple organs, providing exciting insights into fibrotic disease pathogenesis. These include the detailed topographical annotation of pathogenic cell populations and states, detection of transcriptomic perturbations in morphologically normal tissue, characterization of fibrotic and homeostatic niches and their cellular constituents, and in situ interrogation of ligand-receptor interactions within these microenvironments. Together, these powerful readouts enable detailed analysis of fibrosis evolution across time and space.
Collapse
Affiliation(s)
- Prasad Palani Velu
- Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Roxanna E Abhari
- Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Neil C Henderson
- Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4UU, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 1QY, UK
| |
Collapse
|
37
|
Chen Q, Zheng A, Xu X, Shi Z, Yang M, Sun S, Wang L, Wang Y, Zhao H, Xiao Q, Zhang L. Nrf3-Mediated Mitochondrial Superoxide Promotes Cardiomyocyte Apoptosis and Impairs Cardiac Functions by Suppressing Pitx2. Circulation 2025; 151:1024-1046. [PMID: 40099370 DOI: 10.1161/circulationaha.124.070286] [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] [Received: 04/30/2024] [Accepted: 12/31/2024] [Indexed: 03/19/2025]
Abstract
BACKGROUND Myocardial infarction (MI) elicits mitochondria reactive oxygen species (ROS) production and cardiomyocyte (CM) apoptosis. Nrf3 (nuclear factor erythroid 2-related factor 3) has an established role in regulating redox signaling and tissue homeostasis. Here, we aimed to evaluate the role and mechanism of Nrf3 in injury-induced pathological cardiac remodeling. METHODS Global (Nrf3-KO) and CM-specific (Nrf3△CM) Nrf3 knockout mice were subjected to MI or ischemia/reperfusion injury, followed by functional and histopathological analysis. Primary neonatal mouse and rat ventricular myocytes and CMs derived from human induced pluripotent stem cells were used to evaluate the impact of Nrf3 on CM apoptosis and mitochondrial ROS production. Chromatin immunoprecipitation sequencing and immunoprecipitation-mass spectrometry analysis were used to uncover potential targets of Nrf3. MitoParaquat administration and CM-specific adeno-associated virus vectors were used to further confirm the in vivo relevance of the identified signal pathways. RESULTS Nrf3 was expressed mainly in CMs in healthy human hearts, and an increased level of Nrf3 was observed in CMs within the border zone of infarcted human hearts and murine cardiac tissues after MI. Both global and CM-specific Nrf3 knockout significantly decreased injury-induced mitochondrial ROS production, CM apoptosis, and pathological cardiac remodeling, consequently improving cardiac functions. In addition, cardiac-specific Nrf3 overexpression reversed the ameliorative cardiac phenotypes observed in Nrf3-KO mice. Functional studies showed that Nrf3 promoted neonatal mouse ventricular myocyte, neonatal rat ventricular myocyte, and CMs derived from human induced pluripotent stem cell apoptosis by increasing mitochondrial ROS production. Critically, augmenting mitochondrial ROS with MitoParaquat blunted the beneficial effects of Nrf3 deletion on cardiac function and remodeling. Mechanistically, a redox regulator Pitx2 (paired-like homeodomain transcription factor 2) was identified as one of the main target genes of Nrf3. Specifically, Nrf3 binds to Pitx2 promoter, where it increases DNA methylation through recruiting heterogeneous nuclear ribonucleoprotein K and DNA-methyltransferase 1 complex, thereby inhibiting Pitx2 expression. CM-specific knockdown of Pitx2 blunted the beneficial effects of Nrf3 deletion on cardiac function and remodeling, and cardiac-specific Pitx2 overexpression attenuated MI-induced mitochondrial ROS production and CM apoptosis, as well as preserved cardiac functions after MI. CONCLUSIONS Nrf3 promotes injury-induced CM apoptosis and deteriorates cardiac functions by increasing mitochondrial ROS production through suppressing Pitx2 expression. Targeting the Nrf3-Pitx2-mitochondrial ROS signal axis may therefore represent a novel therapeutic approach for MI treatment.
Collapse
Affiliation(s)
- Qishan Chen
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, William Harvey Research, Queen Mary University of London, UK (Q.C., A.Z., Z.S., M.Y., L.W., Q.X.)
| | - Ancheng Zheng
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, William Harvey Research, Queen Mary University of London, UK (Q.C., A.Z., Z.S., M.Y., L.W., Q.X.)
| | - Xiaolei Xu
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
| | - Zhenning Shi
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, William Harvey Research, Queen Mary University of London, UK (Q.C., A.Z., Z.S., M.Y., L.W., Q.X.)
| | - Mei Yang
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, William Harvey Research, Queen Mary University of London, UK (Q.C., A.Z., Z.S., M.Y., L.W., Q.X.)
| | - Shasha Sun
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
| | - Leyu Wang
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, William Harvey Research, Queen Mary University of London, UK (Q.C., A.Z., Z.S., M.Y., L.W., Q.X.)
| | - Yumeng Wang
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
| | - Haige Zhao
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China (H.Z.)
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, William Harvey Research, Queen Mary University of London, UK (Q.C., A.Z., Z.S., M.Y., L.W., Q.X.)
| | - Li Zhang
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China (Q.C., A.Z., X.X., Z.S., M.Y., S.S., L.W., Y.W., L.Z.)
| |
Collapse
|
38
|
Koopmans T, van Rooij E. Molecular gatekeepers of endogenous adult mammalian cardiomyocyte proliferation. Nat Rev Cardiol 2025:10.1038/s41569-025-01145-y. [PMID: 40195566 DOI: 10.1038/s41569-025-01145-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 04/09/2025]
Abstract
Irreversible cardiac fibrosis, cardiomyocyte death and chronic cardiac dysfunction after myocardial infarction pose a substantial global health-care challenge, with no curative treatments available. To regenerate the injured heart, cardiomyocytes must proliferate to replace lost myocardial tissue - a capability that adult mammals have largely forfeited to adapt to the demanding conditions of life. Using various preclinical models, our understanding of cardiomyocyte proliferation has progressed remarkably, leading to the successful reactivation of cell cycle induction in adult animals, with functional recovery after cardiac injury. Central to this success is the targeting of key pathways and structures that drive cardiomyocyte maturation after birth - nucleation and ploidy, sarcomere structure, developmental signalling, chromatin and epigenetic regulation, the microenvironment and metabolic maturation - forming a complex regulatory framework that allows efficient cellular contraction but restricts cardiomyocyte proliferation. In this Review, we explore the molecular pathways underlying these core mechanisms and how their manipulation can reactivate the cell cycle in cardiomyocytes, potentially contributing to cardiac repair.
Collapse
Affiliation(s)
- Tim Koopmans
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, Netherlands.
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands.
| |
Collapse
|
39
|
Fan J, Shen Y, Chen C, Chen X, Yang X, Liu H, Chen R, Liu S, Zhang B, Zhang M, Zhou G, Wang Y, Sun H, Jiang Y, Wei X, Yang T, Liu Y, Tian D, Deng Z, Xu X, Liu X, Tian Z. A large-scale integrated transcriptomic atlas for soybean organ development. MOLECULAR PLANT 2025; 18:669-689. [PMID: 39973009 DOI: 10.1016/j.molp.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/03/2025] [Accepted: 02/15/2025] [Indexed: 02/21/2025]
Abstract
Soybean is one of the most important crops globally, and its production must be significantly increased to meet increasing demand. Elucidating the genetic regulatory networks underlying soybean organ development is essential for breeding elite and resilient varieties to ensure increased soybean production under climate change. An integrated transcriptomic atlas that leverages multiple types of transcriptomics data can facilitate the characterization of temporal-spatial expression patterns of most organ development-related genes and thereby help us to understand organ developmental processes. Here, we constructed a comprehensive, integrated transcriptomic atlas for soybeans, integrating bulk RNA sequencing (RNA-seq) datasets from 314 samples across the soybean life cycle, along with single-nucleus RNA-seq and spatially enhanced resolution omics sequencing datasets from five organs: root, nodule, shoot apex, leaf, and stem. Investigating genes related to organ specificity, blade development, and nodule formation, we demonstrate that the atlas has robust power for exploring key genes involved in organ formation. In addition, we developed a user-friendly panoramic database for the transcriptomic atlas, enabling easy access and queries, which will serve as a valuable resource to significantly advance future soybean functional studies.
Collapse
Affiliation(s)
- Jingwei Fan
- State Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yanting Shen
- State Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Yazhouwan National Laboratory, Sanya 572000, Hainan, China.
| | | | - Xi Chen
- BGI-Beijing, Beijing 102601, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyue Yang
- State Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; College of Advanced Agriculture Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Ruiying Chen
- BGI-Beijing, Beijing 102601, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shulin Liu
- State Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Yazhouwan National Laboratory, Sanya 572000, Hainan, China
| | | | - Min Zhang
- State Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Guoan Zhou
- State Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Yazhouwan National Laboratory, Sanya 572000, Hainan, China
| | - Yu Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Haixi Sun
- BGI-Beijing, Beijing 102601, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuqiang Jiang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaofeng Wei
- China National GeneBank, Shenzhen, Guangdong 518120, China
| | - Tao Yang
- China National GeneBank, Shenzhen, Guangdong 518120, China
| | - Yucheng Liu
- State Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Dongmei Tian
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Ziqing Deng
- BGI-Beijing, Beijing 102601, China; BGI-Shenzhen, Shenzhen 518083, Guangdong, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen 518083, Guangdong, China.
| | - Xin Liu
- BGI-Beijing, Beijing 102601, China; BGI-Shenzhen, Shenzhen 518083, Guangdong, China.
| | - Zhixi Tian
- State Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Yazhouwan National Laboratory, Sanya 572000, Hainan, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
40
|
Qaqorh T, Takahashi Y, Sameshima K, Otani K, Yazawa I, Nishida Y, Tonai K, Fujihara Y, Honda M, Oki S, Ohkawa Y, Thorburn DR, Frazier AE, Takeda A, Ikeda Y, Sakaguchi H, Watanabe T, Fukushima N, Tsukamoto Y, Makita N, Yamaguchi O, Murayama K, Ohtake A, Okazaki Y, Kimura T, Kato H, Inoue H, Matsuoka K, Takashima S, Shintani Y. Atf3 controls transitioning in female mitochondrial cardiomyopathy as identified by spatial and single-cell transcriptomics. SCIENCE ADVANCES 2025; 11:eadq1575. [PMID: 40184463 PMCID: PMC11970478 DOI: 10.1126/sciadv.adq1575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 02/28/2025] [Indexed: 04/06/2025]
Abstract
Oxidative phosphorylation defects result in now intractable mitochondrial diseases (MD) with cardiac involvement markedly affecting prognosis. The mechanisms underlying the transition from compensation to dysfunction in response to metabolic deficiency remain unclear. Here, we used spatially resolved transcriptomics and single-nucleus RNA sequencing (snRNA-seq) on the heart of a patient with mitochondrial cardiomyopathy (MCM), combined with an MCM mouse model with cardiac-specific Ndufs6 knockdown (FS6KD). Cardiomyocytes demonstrated the most heterogeneous expression landscape among cell types caused by metabolic perturbation, and pseudotime trajectory analysis revealed dynamic cellular states transitioning from compensation to severe compromise. This progression coincided with the transient up-regulation of a transcription factor, ATF3. Genetic ablation of Atf3 in FS6KD corroborated its pivotal role, effectively delaying cardiomyopathy progression in a female-specific manner. Our findings highlight a fate-determining role of ATF3 in female MCM progression and that the latest transcriptomic analysis will help decipher the mechanisms underlying MD progression.
Collapse
Affiliation(s)
- Tasneem Qaqorh
- Department of Molecular Pharmacology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
- Department of Medical Biochemistry, Osaka University Graduate School of Frontier Biosciences, Suita, Osaka, Japan
| | - Yusuke Takahashi
- Department of Molecular Pharmacology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Kohei Sameshima
- Department of Molecular Pharmacology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Kentaro Otani
- Department of Molecular Pharmacology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Issei Yazawa
- Department of Molecular Pharmacology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Yuya Nishida
- Department of Molecular Pharmacology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Kohei Tonai
- Department of Molecular Pharmacology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Yoshitaka Fujihara
- Department of Advanced Medical Technologies, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Mizuki Honda
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shinya Oki
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - David R. Thorburn
- Murdoch Children’s Research Institute, Royal Children’s Hospital, and University of Melbourne, Department of Paediatrics, Parkville, Victoria, Australia
- Victorian Clinical Genetics Services, Royal Children’s Hospital, Parkville, Victoria, Australia
| | - Ann E. Frazier
- Murdoch Children’s Research Institute, Royal Children’s Hospital, and University of Melbourne, Department of Paediatrics, Parkville, Victoria, Australia
| | - Atsuhito Takeda
- Department of Pediatrics, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshihiko Ikeda
- Department of Pathology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Heima Sakaguchi
- Department of Pediatric Cardiology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Takuya Watanabe
- Department of Transplant Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Norihide Fukushima
- Department of Transplant Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
- Senri Kinran University, Suita, Osaka, Japan
| | - Yasumasa Tsukamoto
- Department of Transplant Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Naomasa Makita
- Omics Research Center, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
- Department of Cardiology, Sapporo Teishinkai Hospital, Sapporo, Japan
| | - Osamu Yamaguchi
- Omics Research Center, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Kei Murayama
- Department of Metabolism, Chiba Children’s Hospital, Chiba, Japan
- Diagnostics and Therapeutic of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Akira Ohtake
- Department of Pediatrics and Clinical Genomics, Saitama Medical University, Moroyama, Saitama, Japan
| | - Yasushi Okazaki
- Diagnostics and Therapeutic of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Takanari Kimura
- Department of Medical Biochemistry, Osaka University Graduate School of Frontier Biosciences, Suita, Osaka, Japan
| | - Hisakazu Kato
- Department of Medical Biochemistry, Osaka University Graduate School of Frontier Biosciences, Suita, Osaka, Japan
| | - Hijiri Inoue
- Department of Medical Biochemistry, Osaka University Graduate School of Frontier Biosciences, Suita, Osaka, Japan
| | - Ken Matsuoka
- Department of Medical Biochemistry, Osaka University Graduate School of Frontier Biosciences, Suita, Osaka, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Frontier Biosciences, Suita, Osaka, Japan
| | - Yasunori Shintani
- Department of Molecular Pharmacology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| |
Collapse
|
41
|
Zhao Y, Hu Y, Wang Y, Qian H, Zhu C, Dong H, Hao C, Zhang Y, Ji Z, Li X, Chen Y, Xu R, Jiang J, Cao H, Ma G, Chen L. Cardiac fibroblast-derived mitochondria-enriched sEVs regulate tissue inflammation and ventricular remodeling post-myocardial infarction through NLRP3 pathway. Pharmacol Res 2025; 214:107676. [PMID: 40015386 DOI: 10.1016/j.phrs.2025.107676] [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] [Received: 11/25/2024] [Revised: 01/30/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
Resident cardiac fibroblasts (CFs) play crucial roles in sensing injury signals and regulating inflammatory responses post-myocardial infarction (MI). Damaged mitochondria can be transferred extracellularly via various mechanisms, including extracellular vesicles (EVs). In this study, we aimed to investigate whether CFs could transfer damaged mitochondrial components via small EVs (sEVs) and elucidate their role in regulating inflammatory responses post-MI. Left anterior descending coronary artery ligation was performed in mice. Mitochondrial components in sEVs were detected using nanoflow cytometry. Differential protein expression in sEVs from normoxia and normoglycemia CFs (CFs-Nor-sEVs) and CFs post oxygen-glucose deprivation (CFs-OGD-sEVs) was identified using label-free proteomics. CFs-sEVs were co-cultured with mouse bone marrow-derived macrophages (BMDMs) to assess macrophage inflammatory responses. Effects of intramyocardial injection of CFs-sEVs were assessed in MI mice in the absence or presence of NLRP3 inhibitor CY-09. Results demonstrated that mitochondrial components were detected in CFs-derived sEVs post-MI. Damaged mitochondrial components were enriched in CFs-OGD-sEVs (CFs-mt-sEVs), which promoted pro-inflammatory phenotype activation of BMDMs in vitro. Myocardial injection of CFs-mt-sEVs enhanced tissue inflammation, aggravated cardiac dysfunction, and exacerbated maladaptive ventricular remodeling post-MI in vivo. Mechanistically, above effects were achieved via activation of NLRP3 and above effects could be reversed by NLRP3 inhibitor CY-09. This study indicates that CFs could transfer damaged mitochondrial components via the sEVs post-MI, promote macrophage inflammatory activation and exacerbate maladaptive ventricular remodeling post MI by activating NLRP3. Our findings highlight the potential therapeutic effects of inhibiting CFs-mt-sEVs and NLRP3 to improve cardiac function and attenuate ventricular remodeling post-MI.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing 210009, PR China
| | - Ya Hu
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing 210009, PR China
| | - Yifei Wang
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing 210009, PR China
| | - Hao Qian
- Department of Cardiology, Huai 'an No.1 People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Chenxu Zhu
- Institute for Computational Biomedicine - Disease Modeling, RWTH Aachen University, Aachen, Germany
| | - Hongjian Dong
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing 210009, PR China
| | - Chunshu Hao
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing 210009, PR China
| | - Yao Zhang
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing 210009, PR China
| | - Zhenjun Ji
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing 210009, PR China
| | - Xinxin Li
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing 210009, PR China
| | - Yue Chen
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing 210009, PR China
| | - Rongfeng Xu
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing 210009, PR China
| | - Jie Jiang
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing 210009, PR China
| | - Hailong Cao
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing 210009, PR China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing 210009, PR China.
| | - Lijuan Chen
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing 210009, PR China; Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Nanjing 211200, PR China.
| |
Collapse
|
42
|
Tang Z, Alrumaihi F, Alwanian WM, Alharbi HO, Allemailem KS, Alissa M, Alasmari O, Almousa S, Ainsworth T, Chen X. The future of cardiology: Integrating single-cell transcriptomics with multi-omics for enhanced cardiac disease insights. Curr Probl Cardiol 2025; 50:103005. [PMID: 39894239 DOI: 10.1016/j.cpcardiol.2025.103005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
Recent advancements in single-cell transcriptome sequencing (scRNA-seq) have revolutionized our understanding of cellular heterogeneity in cardiovascular diseases, enabling the identification of novel therapeutic targets. This technology allows for high-resolution analysis of gene expression at the single-cell level, revealing the complex dynamics of human heart cell development and the diverse roles of cardiac cell types in health and disease. Despite its transformative potential, current applications of scRNA-seq face limitations, including challenges in data integration and the need for comprehensive multi-omic approaches to fully elucidate the mechanisms underlying cardiovascular pathologies. This review highlights the significant insights gained from scRNA-seq studies in the mammalian heart, emphasizing the importance of integrating spatial transcriptomics and other omics technologies to enhance our understanding of cardiac biology. Furthermore, it addresses the critical research gaps in the field, particularly in the context of personalized medicine and the need for improved methodologies to analyze rare cell populations. By exploring these challenges and opportunities, this review aims to pave the way for innovative diagnostic and therapeutic strategies that can ultimately improve outcomes for patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Zhengchun Tang
- Guangdong Pharmaceutical University, Guangdong Province, 510006, China
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Wanian M Alwanian
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Hajed Obaid Alharbi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Mohammed Alissa
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Omar Alasmari
- Department of Medical Laboratory, Al Kharj Military Industries Corporation Hospital, Al-kharj, Saudi Arabia
| | - Saad Almousa
- Department of Medical Laboratory, Al Kharj Military Industries Corporation Hospital, Al-kharj, Saudi Arabia
| | - Thomas Ainsworth
- Department of Emergency Medicine and Integrative Biosciences Center, Wayne State University, Detroit, MI, United States
| | - Xiangmei Chen
- Guangdong Pharmaceutical University, Guangdong Province, 510006, China.
| |
Collapse
|
43
|
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
|
44
|
Wu H, Lan Q, He YX, Xue JY, Liu H, Zou Y, Liu P, Luo G, Chen MT, Liu MN. Programmed cardiomyocyte death in myocardial infarction. Apoptosis 2025; 30:597-615. [PMID: 39833636 DOI: 10.1007/s10495-025-02075-3] [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] [Accepted: 01/02/2025] [Indexed: 01/22/2025]
Abstract
Cardiovascular disease (CVD) is a leading cause of human mortality worldwide, with patients often at high risk of heart failure (HF) in myocardial infarction (MI), a common form of CVD that results in cardiomyocyte death and myocardial necrosis due to inadequate myocardial perfusion. As terminally differentiated cells, cardiomyocytes possess a severely limited capacity for regeneration, and an excess of dead cardiomyocytes will further stress surviving cells, potentially exacerbating to more extensive heart disease. The article focuses on the relationship between programmed cell death (PCD) of cardiomyocytes, including different forms of apoptosis, necrosis, and autophagy, and MI, as well as the potential application of these mechanisms in the treatment of MI. By gaining a deeper understanding of the mechanisms of cardiomyocyte death, it aims to provide new insights into the prevention and treatment of MI.
Collapse
Affiliation(s)
- Hao Wu
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Qi Lan
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Yi-Xiang He
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Jin-Yi Xue
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Hao Liu
- Department of Pediatrics, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Yuan Zou
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Ping Liu
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Gang Luo
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
| | - Ming-Tai Chen
- Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, 518033, Shenzhen, People's Republic of China.
| | - Meng-Nan Liu
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
| |
Collapse
|
45
|
Schotten U, Goette A, Verheule S. Translation of pathophysiological mechanisms of atrial fibrosis into new diagnostic and therapeutic approaches. Nat Rev Cardiol 2025; 22:225-240. [PMID: 39443702 DOI: 10.1038/s41569-024-01088-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/12/2024] [Indexed: 10/25/2024]
Abstract
Atrial fibrosis is one of the main manifestations of atrial cardiomyopathy, an array of electrical, mechanical and structural alterations associated with atrial fibrillation (AF), stroke and heart failure. Atrial fibrosis can be both a cause and a consequence of AF and, once present, it accelerates the progression of AF. The pathophysiological mechanisms leading to atrial fibrosis are diverse and include stretch-induced activation of fibroblasts, systemic inflammatory processes, activation of coagulation factors and fibrofatty infiltrations. Importantly, atrial fibrosis can occur in different forms, such as reactive and replacement fibrosis. The diversity of atrial fibrosis mechanisms and patterns depends on sex, age and comorbidity profile, hampering the development of therapeutic strategies. In addition, the presence and severity of comorbidities often change over time, potentially causing temporal changes in the mechanisms underlying atrial fibrosis development. This Review summarizes the latest knowledge on the molecular and cellular mechanisms of atrial fibrosis, its association with comorbidities and the sex-related differences. We describe how the various patterns of atrial fibrosis translate into electrophysiological mechanisms that promote AF, and critically appraise the clinical applicability and limitations of diagnostic tools to quantify atrial fibrosis. Finally, we provide an overview of the newest therapeutic interventions under development and discuss relevant knowledge gaps related to the association between clinical manifestations and pathological mechanisms of atrial fibrosis and to the translation of this knowledge to a clinical setting.
Collapse
Affiliation(s)
- Ulrich Schotten
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands.
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands.
| | - Andreas Goette
- Department of Cardiology and Intensive Care Medicine, St. Vincenz Hospital, Paderborn, Germany
- Otto-von-Guericke University, Medical Faculty, Magdeburg, Germany
| | - Sander Verheule
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
46
|
Leblanc FJA, Yiu CHK, Moreira LM, Johnston AM, Mehta N, Kourliouros A, Sayeed R, Nattel S, Reilly S, Lettre G. Single-nucleus multi-omics implicates androgen receptor signaling in cardiomyocytes and NR4A1 regulation in fibroblasts during atrial fibrillation. NATURE CARDIOVASCULAR RESEARCH 2025; 4:433-444. [PMID: 40133651 PMCID: PMC11994452 DOI: 10.1038/s44161-025-00626-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 02/13/2025] [Indexed: 03/27/2025]
Abstract
The dysregulation of gene expression programs in the human atria during persistent atrial fibrillation (AF) is not completely understood. Here, we reanalyze bulk RNA-sequencing datasets from two studies (N = 242) and identified 755 differentially expressed genes in left atrial appendages of individuals with persistent AF and non-AF controls. We combined the bulk RNA-sequencing differentially expressed genes with a left atrial appendage single-nucleus multi-omics dataset to assign genes to specific atrial cell types. We found noncoding genes at the IFNG locus (LINC01479, IFNG-AS1) strongly dysregulated in cardiomyocytes. We defined a gene expression signature potentially driven by androgen receptor signaling in cardiomyocytes from individuals with AF. Cell-type-specific gene expression modules suggested an increase in T cell and a decrease in adipocyte and neuronal cell gene expression in AF. Lastly, we showed that reducing NR4A1 expression, a marker of a poorly characterized human atrial fibroblast subtype, fibroblast activation markers, extracellular matrix remodeling and cell proliferation decreased.
Collapse
MESH Headings
- Humans
- Atrial Fibrillation/genetics
- Atrial Fibrillation/metabolism
- Atrial Fibrillation/pathology
- Atrial Fibrillation/physiopathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Signal Transduction
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Receptors, Androgen/metabolism
- Receptors, Androgen/genetics
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Transcriptome
- Male
- Interferon-gamma/genetics
- Interferon-gamma/metabolism
- Case-Control Studies
- Single-Cell Analysis
- Atrial Appendage/metabolism
- Atrial Appendage/physiopathology
- Atrial Appendage/pathology
- Female
- Gene Expression Regulation
- Gene Expression Profiling
- Middle Aged
- Gene Regulatory Networks
- Cell Proliferation
- Multiomics
Collapse
Affiliation(s)
- Francis J A Leblanc
- Montreal Heart Institute, Montreal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Chi Him Kendrick Yiu
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Lucia M Moreira
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Aaron M Johnston
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Neelam Mehta
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Antonios Kourliouros
- Cardiothoracic Surgery, Oxford Heart Centre, John Radcliffe Hospital, Oxford, UK
| | - Rana Sayeed
- Cardiothoracic Surgery, Oxford Heart Centre, John Radcliffe Hospital, Oxford, UK
| | - Stanley Nattel
- Montreal Heart Institute, Montreal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montréal, Quebec, Canada
- IHU Liryc and Fondation Bordeaux Université, Bordeaux, France
- Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Essen, Germany
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK.
| | - Guillaume Lettre
- Montreal Heart Institute, Montreal, Quebec, Canada.
- Department of Medicine, Université de Montréal, Montréal, Quebec, Canada.
| |
Collapse
|
47
|
Hayes JB, Bainbridge AM, Burnette DT. Alpha-actinin-1 stabilizes focal adhesions to facilitate sarcomere assembly in cardiac myocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.645933. [PMID: 40196508 PMCID: PMC11974845 DOI: 10.1101/2025.03.28.645933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Cardiac sarcomere assembly is a highly orchestrated process requiring integration between intracellular contractile components and extracellular adhesions. While α-actinin-2 (ACTN2) is well known for its structural role at Z-discs, the function of the "non-muscle" paralog α-actinin-1 (ACTN1) in cardiomyocytes remains unclear. Using human induced pluripotent stem cell-derived cardiac myocytes (hiCMs), we demonstrate that ACTN1 is essential for sarcomere assembly. siRNA-mediated depletion of ACTN1 disrupted Z-line formation and impaired sarcomere organization, defects that were rescued by exogenous ACTN1 but not ACTN2, revealing non-redundant functions. Unlike ACTN2, ACTN1 localized predominantly to focal adhesions and was required for adhesion maturation, as evidenced by reduced adhesion size and number following ACTN1 depletion. Live-cell imaging of vinculin dynamics showed decreased stability of adhesion-associated vinculin in ACTN1-deficient cells, whereas paxillin dynamics were unaffected. These results suggest that ACTN1 stabilizes focal adhesions to promote effective force transmission during sarcomere assembly.
Collapse
Affiliation(s)
- James B Hayes
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Anna M Bainbridge
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
- University of Tennessee, Knoxville, TN, USA
| | - Dylan T Burnette
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| |
Collapse
|
48
|
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] [Download PDF] [Figures] [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
|
49
|
Arjamaa O. Hypoxia in myocardial infarction and natriuretic peptides. Open Heart 2025; 12:e003130. [PMID: 40122569 PMCID: PMC11934622 DOI: 10.1136/openhrt-2024-003130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Mechanical stress on the heart is commonly considered the sole stimulus explaining the synthesis and release of circulating natriuretic peptides and their derivatives. While one of the most critical paradigms in cardiology is that mechanical load increases oxygen consumption, clinical studies on these peptides have neglected the relationship between mechanical stress and oxygen metabolism. At the cellular level, cardiac myocytes have a ubiquitous oxygen-sensing pathway mediated by a nuclear transcription factor, the hypoxia-inducible factor (HIF). Published studies indicate that the human myocardium starts expressing HIF during infarction. In myocardial cell cultures, natriuretic peptides are synthesised and released under hypoxic conditions through immediate and sufficient actions of HIF. CONCLUSION Myocardial oxygen metabolism directly regulates the plasma levels of natriuretic peptides in heart diseases. The function of oxygen gradients should be correlated with circulating natriuretic peptides to achieve better sensitivity in plasma measurements of natriuretic peptides in myocardial infarction.
Collapse
Affiliation(s)
- Olli Arjamaa
- University of Turku Biodiversity Unit, Turku, Finland
| |
Collapse
|
50
|
Mulvey JF, Meyer EL, Svenningsen MS, Lundby A. Integrating -Omic Technologies across Modality, Space, and Time to Decipher Remodeling in Cardiac Disease. Curr Cardiol Rep 2025; 27:74. [PMID: 40116972 PMCID: PMC11928419 DOI: 10.1007/s11886-025-02226-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/11/2025] [Indexed: 03/23/2025]
Abstract
PURPOSE OF REVIEW Despite significant efforts to understand pathophysiological processes underlying cardiac diseases, the molecular causes for the most part remain unresolved. Rapid advancements in -omics technologies, and their application in cardiac research, offer new insight into cardiac remodeling in disease states. This review aims to provide an accessible overview of recent advances in omics approaches for studying cardiac remodeling, catering to readers without extensive prior expertise. RECENT FINDINGS We provide a methodologically focused overview of current methods for performing transcriptomics and proteomics, including their extensions for single-cell and spatial measurements. We discuss approaches to integrate data across modalities, resolutions and time. Key recent applications within the cardiac field are highlighted. Each -omics modality can provide insight, yet each existing experimental method has technical or conceptual limitations. Integrating data across multiple modalities can leverage strengths and mitigate weaknesses, ultimately enhancing our understanding of cardiac pathophysiology.
Collapse
Affiliation(s)
- John F Mulvey
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emily L Meyer
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel Skjoldan Svenningsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alicia Lundby
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|