1
|
Mitra A, Mandal S, Banerjee K, Ganguly N, Sasmal P, Banerjee D, Gupta S. Cardiac Regeneration in Adult Zebrafish: A Review of Signaling and Metabolic Coordination. Curr Cardiol Rep 2025; 27:15. [PMID: 39792206 DOI: 10.1007/s11886-024-02162-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/20/2024] [Indexed: 01/12/2025]
Abstract
PURPOSE OF REVIEW This review investigates how post-injury cellular signaling and energy metabolism are two pivotal points in zebrafish's cardiomyocyte cell cycle re-entry and proliferation. It seeks to highlight the probable mechanism of action in proliferative cardiomyocytes compared to mammals and identify gaps in the current understanding of metabolic regulation of cardiac regeneration. RECENT FINDINGS Metabolic substrate changes after birth correlate with reduced cardiomyocyte proliferation in mammals. Unlike adult mammalian hearts, zebrafish can regenerate cardiomyocytes by re-entering the cell cycle, characterized by a metabolic switch from oxidative metabolism to increased glycolysis. Zebrafish provide a valuable model for studying metabolic regulation during cell cycle re-entry and cardiac regeneration. Proliferative cardiomyocytes have upregulated Notch, hippo, and Wnt signaling and decreased ROS generation, DNA damage in different zebrafish cardiac regeneration models. Understanding the correlation between metabolic switches during cell cycle re-entry of already differentiated zebrafish cardiomyocytes is being increasingly recognized as a critical factor in heart regeneration. Zebrafish studies provide insights into metabolic adaptations during heart regeneration, emphasizing the importance of a metabolic switch. However, there are mechanistic gaps, and extensive studies are required to aid in formulating therapeutic strategies for cardiac regenerative medicine.
Collapse
Affiliation(s)
- Arkadeep Mitra
- Department of Zoology, City College, 102/1, Raja Rammohan Sarani, Kolkata, 700009, West Bengal, India
| | - Subhadeep Mandal
- Department of Zoology, Trivenidevi Bhalotia College (Affiliated to Kazi Nazrul University), College Para Rd, Raniganj, 713347, West Bengal, India
| | - Kalyan Banerjee
- Department of Zoology, Trivenidevi Bhalotia College (Affiliated to Kazi Nazrul University), College Para Rd, Raniganj, 713347, West Bengal, India
| | - Nilanjan Ganguly
- Department of Zoology, Trivenidevi Bhalotia College (Affiliated to Kazi Nazrul University), College Para Rd, Raniganj, 713347, West Bengal, India
| | - Pramit Sasmal
- Department of Zoology, Trivenidevi Bhalotia College (Affiliated to Kazi Nazrul University), College Para Rd, Raniganj, 713347, West Bengal, India
| | - Durba Banerjee
- Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican St, Seattle, WA, 98109, USA.
| | - Shreyasi Gupta
- Department of Zoology, Trivenidevi Bhalotia College (Affiliated to Kazi Nazrul University), College Para Rd, Raniganj, 713347, West Bengal, India.
| |
Collapse
|
2
|
Bouwman M, de Bakker DEM, Honkoop H, Giovou AE, Versteeg D, Boender AR, Nguyen PD, Slotboom M, Colquhoun D, Vigil-Garcia M, Kooijman L, Janssen R, Hooijkaas IB, Günthel M, Visser KJ, Klerk M, Zentilin L, Giacca M, Kaslin J, Boink GJJ, van Rooij E, Christoffels VM, Bakkers J. Cross-species comparison reveals that Hmga1 reduces H3K27me3 levels to promote cardiomyocyte proliferation and cardiac regeneration. NATURE CARDIOVASCULAR RESEARCH 2025; 4:64-82. [PMID: 39747457 PMCID: PMC11738996 DOI: 10.1038/s44161-024-00588-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 11/26/2024] [Indexed: 01/04/2025]
Abstract
In contrast to adult mammalian hearts, the adult zebrafish heart efficiently replaces cardiomyocytes lost after injury. Here we reveal shared and species-specific injury response pathways and a correlation between Hmga1, an architectural non-histone protein, and regenerative capacity, as Hmga1 is required and sufficient to induce cardiomyocyte proliferation and required for heart regeneration. In addition, Hmga1 was shown to reactivate developmentally silenced genes, likely through modulation of H3K27me3 levels, poising them for a pro-regenerative gene program. Furthermore, AAV-mediated Hmga1 expression in injured adult mouse hearts led to controlled cardiomyocyte proliferation in the border zone and enhanced heart function, without cardiomegaly and adverse remodeling. Histone modification mapping in mouse border zone cardiomyocytes revealed a similar modulation of H3K27me3 marks, consistent with findings in zebrafish. Our study demonstrates that Hmga1 mediates chromatin remodeling and drives a regenerative program, positioning it as a promising therapeutic target to enhance cardiac regeneration after injury.
Collapse
Affiliation(s)
- Mara Bouwman
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dennis E M de Bakker
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Hessel Honkoop
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Alexandra E Giovou
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Danielle Versteeg
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Arie R Boender
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- PacingCure BV, Amsterdam, The Netherlands
| | - Phong D Nguyen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
- Institut Curie, Université PSL, CNRS UMR3215, INSERM U934, Paris, France
| | - Merel Slotboom
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Daniel Colquhoun
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Marta Vigil-Garcia
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lieneke Kooijman
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rob Janssen
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Ingeborg B Hooijkaas
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Marie Günthel
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Kimberly J Visser
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mischa Klerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Lorena Zentilin
- International Centre for Genetic Engineering and Biotechnology (ICGEB), University of Trieste, Trieste, Italy
| | - Mauro Giacca
- International Centre for Genetic Engineering and Biotechnology (ICGEB), University of Trieste, Trieste, Italy
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Jan Kaslin
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Gerard J J Boink
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- PacingCure BV, Amsterdam, The Netherlands
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Jeroen Bakkers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands.
- Department of Pediatric Cardiology, Division of Pediatrics, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
3
|
Sweeney M, O'Fee K, Villanueva-Hayes C, Rahman E, Lee M, Tam CN, Pascual-Navarro E, Maatz H, Lindberg EL, Vanezis K, Ramachandra CJ, Andrew I, Jennings ER, Lim WW, Widjaja AA, Carling D, Hausenloy DJ, Hübner N, Barton PJR, Cook SA. Interleukin 11 therapy causes acute left ventricular dysfunction. Cardiovasc Res 2024; 120:2220-2235. [PMID: 39383190 DOI: 10.1093/cvr/cvae224] [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: 06/25/2024] [Revised: 08/13/2024] [Accepted: 09/15/2024] [Indexed: 10/11/2024] Open
Abstract
AIMS Interleukin 11 (IL11) was initially thought important for platelet production, which led to recombinant IL11 being developed as a drug to treat thrombocytopenia. IL11 was later found to be redundant for haematopoiesis, and its use in patients is associated with unexplained and severe cardiac side effects. Here, we aim to identify, for the first time, direct cardiomyocyte toxicities associated with IL11, which was previously believed cardioprotective. METHODS AND RESULTS We injected recombinant mouse lL11 (rmIL11) into mice and studied its molecular effects in the heart using immunoblotting, qRT-PCR, bulk RNA-seq, single nuclei RNA-seq (snRNA-seq), and assay for transposase-accessible chromatin with sequencing (ATAC-seq). The physiological impact of IL11 was assessed by echocardiography in vivo and using cardiomyocyte contractility assays in vitro. To determine the activity of IL11 specifically in cardiomyocytes, we made two cardiomyocyte-specific Il11ra1 knockout (CMKO) mouse models using either AAV9-mediated and Tnnt2-restricted (vCMKO) or Myh6 (m6CMKO) Cre expression and an Il11ra1 floxed mouse strain. In pharmacologic studies, we studied the effects of JAK/STAT inhibition on rmIL11-induced cardiac toxicities. Injection of rmIL11 caused acute and dose-dependent impairment of left ventricular ejection fraction (saline: 62.4% ± 1.9; rmIL11: 32.6% ± 2.9, P < 0.001, n = 5). Following rmIL11 injection, myocardial STAT3 and JNK phosphorylation were increased and bulk RNA-seq revealed up-regulation of pro-inflammatory pathways (TNFα, NFκB, and JAK/STAT) and perturbed calcium handling. snRNA-seq showed rmIL11-induced expression of stress factors (Ankrd1, Ankrd23, Xirp2), activator protein-1 (AP-1) transcription factor genes, and Nppb in the cardiomyocyte compartment. Following rmIL11 injection, ATAC-seq identified the Ankrd1 and Nppb genes and loci enriched for stress-responsive, AP-1 transcription factor binding sites. Cardiomyocyte-specific effects were examined in vCMKO and m6CMKO mice, which were both protected from rmIL11-induced left ventricular impairment and molecular pathobiologies. In mechanistic studies, inhibition of JAK/STAT signalling with either ruxolitinib or tofacitinib prevented rmIL11-induced cardiac dysfunction. CONCLUSIONS Injection of IL11 directly activates IL11RA/JAK/STAT3 in cardiomyocytes to cause acute heart failure. Our data overturn the earlier assumption that IL11 is cardioprotective and explain the serious cardiac side effects associated with IL11 therapy.
Collapse
MESH Headings
- Animals
- Interleukin-11/metabolism
- Interleukin-11/genetics
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Ventricular Function, Left/drug effects
- Mice, Knockout
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/genetics
- Ventricular Dysfunction, Left/chemically induced
- Ventricular Dysfunction, Left/prevention & control
- Signal Transduction
- Disease Models, Animal
- Mice, Inbred C57BL
- STAT3 Transcription Factor/metabolism
- STAT3 Transcription Factor/genetics
- Janus Kinases/metabolism
- Janus Kinase Inhibitors/pharmacology
- Acute Disease
- Male
- Myocardial Contraction/drug effects
- Nitriles/pharmacology
- Recombinant Proteins/metabolism
- Cells, Cultured
- Pyrimidines/pharmacology
- Pyrazoles/pharmacology
- Phosphorylation
- Mice
- Protein Kinase Inhibitors/pharmacology
Collapse
Affiliation(s)
- Mark Sweeney
- MRC-Laboratory of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Katie O'Fee
- MRC-Laboratory of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Chelsie Villanueva-Hayes
- MRC-Laboratory of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Ekhlas Rahman
- MRC-Laboratory of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Michael Lee
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Chung Nga Tam
- MRC-Laboratory of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Eneko Pascual-Navarro
- MRC-Laboratory of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Henrike Maatz
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13092 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Eric L Lindberg
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13092 Berlin, Germany
| | - Konstantinos Vanezis
- MRC-Laboratory of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Chrishan J Ramachandra
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore 169609, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Ivan Andrew
- MRC-Laboratory of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Emma R Jennings
- MRC-Laboratory of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Wei-Wen Lim
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore 169609, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Anissa A Widjaja
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - David Carling
- MRC-Laboratory of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Derek J Hausenloy
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore 169609, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- The Hatter Cardiovascular Institute, University College London, London WC1E 6HX, UK
| | - Norbert Hübner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13092 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Charite, Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Paul J R Barton
- MRC-Laboratory of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
- Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London SW3 6NP, UK
| | - Stuart A Cook
- MRC-Laboratory of Medical Sciences, Hammersmith Hospital Campus, London W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore 169609, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| |
Collapse
|
4
|
Forman-Rubinsky R, Feng W, Schlegel BT, Paul A, Zuppo D, Kedziora K, Stoltz D, Watkins S, Rajasundaram D, Li G, Tsang M. Cited4a limits cardiomyocyte dedifferentiation and proliferation during zebrafish heart regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.05.626917. [PMID: 39713454 PMCID: PMC11661073 DOI: 10.1101/2024.12.05.626917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Cardiac regeneration involves the interplay of complex interactions between many different cell types, including cardiomyocytes. The exact mechanism that enables cardiomyocytes to undergo dedifferentiation and proliferation to replace lost cells has been intensely studied. Here we report a single nuclear RNA sequencing profile of the injured zebrafish heart and identify distinct cardiomyocyte populations in the injured heart. These cardiomyocyte populations have diverse functions, including stress response, myofibril assembly, proliferation and contraction. The contracting cardiomyocyte population also involves the activation of maturation pathways as an early response to injury. This intriguing finding suggests that constant maintenance of a distinctive terminally differentiated cardiomyocyte population is important for cardiac function during regeneration. To test this hypothesis, we determined that cited4a, a p300/CBP transcriptional coactivator, is induced after injury in the mature cardiomyocyte population. Moreover, loss-of-cited4a mutants presented increased dedifferentiation, proliferation and accelerated heart regeneration. Thus, suppressing cardiomyocyte maturation pathway activity in injured hearts could be an approach to promote heart regeneration.
Collapse
Affiliation(s)
- Rachel Forman-Rubinsky
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Integrative Organ Systems, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Wei Feng
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Integrative Organ Systems, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Brent T Schlegel
- Department of Pediatrics, Division of Health Informatics, Children's Hospital of Pittsburgh, Pittsburgh, PA
| | - Angela Paul
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Integrative Organ Systems, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Daniel Zuppo
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Current address: Department of Medicine, University of Rochester Medical Center Rochester, NY
| | - Katarzyna Kedziora
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Biological Imaging, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Donna Stoltz
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Biological Imaging, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Simon Watkins
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Biological Imaging, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, Division of Health Informatics, Children's Hospital of Pittsburgh, Pittsburgh, PA
| | - Guang Li
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Integrative Organ Systems, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Michael Tsang
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Integrative Organ Systems, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| |
Collapse
|
5
|
Chen Y, Liang R, Li Y, Jiang L, Ma D, Luo Q, Song G. Chromatin accessibility: biological functions, molecular mechanisms and therapeutic application. Signal Transduct Target Ther 2024; 9:340. [PMID: 39627201 PMCID: PMC11615378 DOI: 10.1038/s41392-024-02030-9] [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: 03/11/2024] [Revised: 08/04/2024] [Accepted: 10/17/2024] [Indexed: 12/06/2024] Open
Abstract
The dynamic regulation of chromatin accessibility is one of the prominent characteristics of eukaryotic genome. The inaccessible regions are mainly located in heterochromatin, which is multilevel compressed and access restricted. The remaining accessible loci are generally located in the euchromatin, which have less nucleosome occupancy and higher regulatory activity. The opening of chromatin is the most important prerequisite for DNA transcription, replication, and damage repair, which is regulated by genetic, epigenetic, environmental, and other factors, playing a vital role in multiple biological progresses. Currently, based on the susceptibility difference of occupied or free DNA to enzymatic cleavage, solubility, methylation, and transposition, there are many methods to detect chromatin accessibility both in bulk and single-cell level. Through combining with high-throughput sequencing, the genome-wide chromatin accessibility landscape of many tissues and cells types also have been constructed. The chromatin accessibility feature is distinct in different tissues and biological states. Research on the regulation network of chromatin accessibility is crucial for uncovering the secret of various biological processes. In this review, we comprehensively introduced the major functions and mechanisms of chromatin accessibility variation in different physiological and pathological processes, meanwhile, the targeted therapies based on chromatin dynamics regulation are also summarized.
Collapse
Affiliation(s)
- Yang Chen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Rui Liang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Yong Li
- Hepatobiliary Pancreatic Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Lingli Jiang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Di Ma
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China.
| |
Collapse
|
6
|
Wang ZY, Mehra A, Wang QC, Gupta S, Ribeiro da Silva A, Juan T, Günther S, Looso M, Detleffsen J, Stainier DYR, Marín-Juez R. flt1 inactivation promotes zebrafish cardiac regeneration by enhancing endothelial activity and limiting the fibrotic response. Development 2024; 151:dev203028. [PMID: 39612288 PMCID: PMC11634031 DOI: 10.1242/dev.203028] [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: 05/08/2024] [Accepted: 10/22/2024] [Indexed: 12/01/2024]
Abstract
VEGFA administration has been explored as a pro-angiogenic therapy for cardiovascular diseases including heart failure for several years, but with little success. Here, we investigate a different approach to augment VEGFA bioavailability: by deleting the VEGFA decoy receptor VEGFR1 (also known as FLT1), one can achieve more physiological VEGFA concentrations. We find that after cryoinjury, zebrafish flt1 mutant hearts display enhanced coronary revascularization and endocardial expansion, increased cardiomyocyte dedifferentiation and proliferation, and decreased scarring. Suppressing Vegfa signaling in flt1 mutants abrogates these beneficial effects of flt1 deletion. Transcriptomic analyses of cryoinjured flt1 mutant hearts reveal enhanced endothelial MAPK/ERK signaling and downregulation of the transcription factor gene egr3. Using newly generated genetic tools, we observe egr3 upregulation in the regenerating endocardium, and find that Egr3 promotes myofibroblast differentiation. These data indicate that with enhanced Vegfa bioavailability, the endocardium limits myofibroblast differentiation via egr3 downregulation, thereby providing a more permissive microenvironment for cardiomyocyte replenishment after injury.
Collapse
Affiliation(s)
- Zhen-Yu Wang
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
| | - Armaan Mehra
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
| | - Qian-Chen Wang
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
| | - Savita Gupta
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
| | - Agatha Ribeiro da Silva
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
| | - Thomas Juan
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
| | - Stefan Günther
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Mario Looso
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
- Bioinformatics Core Unit (BCU), Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Jan Detleffsen
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
- Bioinformatics Core Unit (BCU), Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Didier Y. R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
| | - Rubén Marín-Juez
- Centre Hospitalier Universitaire Sainte-Justine Research Center, 3175 Chemin de la Côte-Sainte-Catherine, H3T 1C5 Montréal, QC, Canada
- Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, H3T 1J4 Montréal, QC, Canada
| |
Collapse
|
7
|
Zhang L, Zhou J. Zebrafish: A smart tool for heart disease research. JOURNAL OF FISH BIOLOGY 2024; 105:1487-1500. [PMID: 37824489 DOI: 10.1111/jfb.15585] [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: 07/22/2023] [Revised: 09/07/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
The increasing prevalence of heart disease poses a significant threat to human survival and safety. However, the current treatments available for heart disease are quite limited. Therefore, it is important to utilize suitable animal models that can accurately simulate the physiological characteristics of heart disease. This would help improve our understanding of this disease and aid in the development of new treatment methods and drugs. Zebrafish heart not only exhibits similarities to mammalian hearts, but they also share ~70% of homologous genes with humans. Utilizing zebrafish as an alternative to expensive and time-consuming mammalian models offers numerous advantages. Zebrafish models can be easily established and maintained, and compound screening and genetic methods allow for the development of various economical and easily controlled zebrafish and zebrafish embryonic heart disease models in a short period of time. Consequently, zebrafish have become a powerful tool for exploring the pathological mechanisms of heart disease and identifying new effective genes. In this review, we summarize recent studies on different zebrafish models of heart disease. We also describe the techniques and protocols used to develop zebrafish models of myocardial infarction, heart failure, and congenital heart disease, including surgical procedures, forward and reverse genetics, and drug and combination screening. This review aims to promote the utilization of zebrafish models in investigating diverse pathological mechanisms of heart disease, enhancing our knowledge and comprehension of heart disease, and offering novel insights and objectives for exploring the prevention and treatment of heart disease.
Collapse
Affiliation(s)
- Lantian Zhang
- Education Branch, Chongqing Publishing Group, Chongqing, China
| | - Jinrun Zhou
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, China
| |
Collapse
|
8
|
Zhao Y, Liang J, Liu X, Li H, Chang C, Gao P, Du F, Zhang R. Tcap deficiency impedes striated muscle function and heart regeneration with elevated ROS and autophagy. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167485. [PMID: 39226992 DOI: 10.1016/j.bbadis.2024.167485] [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: 08/07/2024] [Accepted: 08/26/2024] [Indexed: 09/05/2024]
Abstract
Telethonin/titin-cap (TCAP) encodes a Z-disc protein that plays important roles in sarcomere/T-tubule interactions, stretch-sensing and signaling. Mutations in TCAP are associated with muscular dystrophy and cardiomyopathy; however, the complete etiology and its roles in myocardial infarction and regeneration are not fully understood. Here, we generated tcap gene knockout zebrafish with CRISPR/Cas9 technology and observed muscular dystrophy-like phenotypes and abnormal mitochondria in skeletal muscles. The stretch-sensing ability was inhibited in tcap-/- mutants. Moreover, Tcap deficiency led to alterations in cardiac morphology and function as well as increases in reactive oxygen species (ROS) and mitophagy. In addition, the cardiac regeneration and cardiomyocyte proliferation ability of tcap-/- mutants were impaired, but these impairments could be rescued by supplementation with ROS scavengers or autophagy inhibitors. Overall, our study demonstrates the essential roles of Tcap in striated muscle function and heart regeneration. Additionally, elevations in ROS and autophagy may account for the phenotypes resulting from Tcap deficiency and could serve as novel therapeutic targets for muscular dystrophy and cardiomyopathy.
Collapse
Affiliation(s)
- Yan Zhao
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China.
| | - Jieling Liang
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Xuan Liu
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Huicong Li
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Cheng Chang
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Peng Gao
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Fen Du
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China.
| | - Ruilin Zhang
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China.
| |
Collapse
|
9
|
Guo X, Zhang M, Xu Z, Yu Y, Shen Z. CRISPR/Cas9-mediated generation of AP-1 activity reporter cell line in human embryonic stem cell (WAe007-A-5). Stem Cell Res 2024; 81:103557. [PMID: 39276528 DOI: 10.1016/j.scr.2024.103557] [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] [Received: 07/18/2024] [Revised: 08/14/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024] Open
Abstract
Activator protein 1 (AP-1) is involved in cell fate determination and function. To monitor the AP-1 activity, we cloned a AP-1 binding sites fragment into the upstream of minimal TATA-box promoter, then a luciferase-GFP reporter (LuciGFP) was designated to the downstream of the promoter. With CRISPR/Cas9, the AP-1-LuciGFP reporter was introduced into AAVS1 locus, a safe harbor for gene editing. Thus, this AP-1-LuciGFP reporter cell line could be subjected to monitor the AP-1 activity during the cell differentiation, cell fate transition and disease modeling.
Collapse
Affiliation(s)
- Xingyou Guo
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou 215123, Jiangsu, PR China; Suzhou Medical College, Soochow University, Suzhou 215123, Jiangsu, PR China; Department of Vascular Surgery, Suqian First Hospital, Suqian 223800, Jiangsu, PR China
| | - Meng Zhang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou 215123, Jiangsu, PR China; Suzhou Medical College, Soochow University, Suzhou 215123, Jiangsu, PR China
| | - Zhanglei Xu
- Suzhou Medical College, Soochow University, Suzhou 215123, Jiangsu, PR China; School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, Jiangsu, PR China
| | - You Yu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou 215123, Jiangsu, PR China; Suzhou Medical College, Soochow University, Suzhou 215123, Jiangsu, PR China.
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou 215123, Jiangsu, PR China; Suzhou Medical College, Soochow University, Suzhou 215123, Jiangsu, PR China.
| |
Collapse
|
10
|
Gu J, Chen X, Luo Z, Li R, Xu Q, Liu M, Liu X, Liu Y, Jiang S, Zou M, Ling S, Liu S, Liu N. Cardiomyocyte-derived exosomes promote cardiomyocyte proliferation and neonatal heart regeneration. FASEB J 2024; 38:e70186. [PMID: 39560071 DOI: 10.1096/fj.202400737rr] [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: 04/03/2024] [Revised: 10/21/2024] [Accepted: 11/04/2024] [Indexed: 11/20/2024]
Abstract
Heart regeneration was mainly achieved by intrinsic capacity. Exosomes are crucial in cardiovascular disease, yet their involvement in myocardial regeneration remains underexplored. To understand the role of cardiomyocyte-derived exosomes (CM-Exos) in heart regeneration. We established mouse models of myocardial infarction and apical resection in neonates to investigate the potential benefits of exosomes in response to injury. Rab27a knockout (KO) mice were constructed as an exosome decrease model. Distinct fibrosis appears in the infarcted and resection area in the KO mice 21 days after heart injury. The proliferation marker pH 3, Ki67, and Aurora B were detected 3 days after surgery, which decreased in KO mice compared to WT mice. Intravenous injection of CM-Exos increased cardiomyocyte proliferation and partially restored heart function in KO mice. Rab27a knockdown in vitro reduced the expression of pH 3, Ki67, and Aurora B positive cardiomyocytes. However, the supplementation of CM-Exos increased the proliferation of cardiomyocytes. Exosomal miRNA sequencing was subsequently applied, and miR-21-5p was a promising candidate that promoted cardiomyocyte proliferation through its target genes Spry-1 and PDCD4. Intravenous injection of miR-21-5p exhibited similar proliferative effects as CM-Exos. Our results indicate that CM-Exos promotes cardiomyocyte cycle reentry by delivering miR-21-5p, highlighting the endogenous factors of myocardial regeneration.
Collapse
Affiliation(s)
- Jielei Gu
- Guangdong Key Laboratory of Vascular Diseases, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xuke Chen
- Guangdong Key Laboratory of Vascular Diseases, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhenyu Luo
- Guangdong Key Laboratory of Vascular Diseases, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Rongxue Li
- Guangdong Key Laboratory of Vascular Diseases, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qiong Xu
- Guangdong Key Laboratory of Vascular Diseases, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Mingke Liu
- Guangdong Key Laboratory of Vascular Diseases, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaolin Liu
- Guangdong Key Laboratory of Vascular Diseases, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yajing Liu
- Guangdong Key Laboratory of Vascular Diseases, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Siqin Jiang
- Guangdong Key Laboratory of Vascular Diseases, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Min Zou
- Guangdong Key Laboratory of Vascular Diseases, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Sisi Ling
- Guangdong Key Laboratory of Vascular Diseases, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shiming Liu
- Guangdong Key Laboratory of Vascular Diseases, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ningning Liu
- Guangdong Key Laboratory of Vascular Diseases, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
11
|
Zhao WJ, Qian Y, Zhang YF, Yang AH, Cao JX, Qian HY, Liu Y, Zhu WZ. Endothelial FOSL1 drives angiotensin II-induced myocardial injury via AT1R-upregulated MYH9. Acta Pharmacol Sin 2024:10.1038/s41401-024-01410-9. [PMID: 39592734 DOI: 10.1038/s41401-024-01410-9] [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: 05/15/2024] [Accepted: 10/09/2024] [Indexed: 11/28/2024] Open
Abstract
Vascular remodeling represents a pathological basis for myocardial pathologies, including myocardial hypertrophy and myocardial infarction, which can ultimately lead to heart failure. The molecular mechanism of angiotensin II (Ang II)-induced vascular remodeling following myocardial infarction reperfusion is complex and not yet fully understood. In this study, we examined the effect of Ang II infusion on cardiac vascular remodeling in mice. Single-cell sequencing showed Ang II induced cytoskeletal pathway enrichment and that FOS like-1 (FOSL1) affected mouse cardiac endothelial dysfunction by pseudotime analysis. Myosin heavy chain 9 (MYH9) was predominantly expressed in primary cardiac endothelial cells. The Ang II type I receptor blocker telmisartan and the protein kinase C inhibitor staurosporine suppressed Ang II-induced upregulation of MYH9 and FOSL1 phosphorylation in human umbilical vein endothelial cells. Silencing MYH9 abolished Ang II-mediated inhibition of angiogenesis in human umbilical vein endothelial cells, and attenuated AngII-induced vascular hyperpermeability. We found that FOSL1 directly bound to the MYH9 promoter and thus activated transcription of MYH9 by the dual luciferase reporter and chromatin immunoprecipitation assays, leading to vascular dysfunction. In vivo, 6 weeks after injecting adeno-associated virus-ENT carrying the TEK tyrosine kinase (tie) promoter-driven short hairpin RNA for silencing FOSL1 (AAV-tie-shFOSL1), cardiac function represented by the ejection fraction and fractional shortening was improved, myocardial fibrosis was decreased, protein levels of phosphorylated FOSL1, MYH9, and collagen type I alpha were reduced, and cardiac vascular density was recovered in mice with endothelial Fosl1-specific knockdown in Ang II-infused mice. In ischemia-reperfusion mice, AAV-shFosl1 mice had a reduced infarct size and preserved cardiac function compared with control AAV mice. Our findings suggest a critical role of the FOSL1/MYH9 axis in hindering Ang II-induced vascular remodeling, and we identified FOSL1 as a potential therapeutic target in endothelial cell injuries induced by myocardial ischemia-reperfusion.
Collapse
Affiliation(s)
- Wen-Jing Zhao
- Department of Pharmacology, School of Medicine and School of Pharmacy Nantong University, Nantong, 226001, China
- Cancer Research Center Nantong, Nantong Tumor Hospital and Tumor Hospital Affiliated to Nantong University, Nantong, 226006, China
| | - Yi Qian
- Department of Pharmacology, School of Medicine and School of Pharmacy Nantong University, Nantong, 226001, China
| | - Yi-Feng Zhang
- Department of Pharmacology, School of Medicine and School of Pharmacy Nantong University, Nantong, 226001, China
| | - Ai-Hua Yang
- Department of Pharmacology, School of Medicine and School of Pharmacy Nantong University, Nantong, 226001, China
| | - Jia-Xin Cao
- Department of Pharmacology, School of Medicine and School of Pharmacy Nantong University, Nantong, 226001, China
| | - Hong-Yan Qian
- Cancer Research Center Nantong, Nantong Tumor Hospital and Tumor Hospital Affiliated to Nantong University, Nantong, 226006, China
| | - Yi Liu
- Department of Pharmacology, School of Medicine and School of Pharmacy Nantong University, Nantong, 226001, China
| | - Wei-Zhong Zhu
- Department of Pharmacology, School of Medicine and School of Pharmacy Nantong University, Nantong, 226001, China.
| |
Collapse
|
12
|
Hofmann C, Serafin A, Schwerdt OM, Fischer J, Sicklinger F, Younesi FS, Byrne NJ, Meyer IS, Malovrh E, Sandmann C, Jürgensen L, Kamuf-Schenk V, Stroh C, Löwenthal Z, Finke D, Boileau E, Beisaw A, Bugger H, Rettel M, Stein F, Katus HA, Jakobi T, Frey N, Leuschner F, Völkers M. Transient Inhibition of Translation Improves Cardiac Function After Ischemia/Reperfusion by Attenuating the Inflammatory Response. Circulation 2024; 150:1248-1267. [PMID: 39206545 PMCID: PMC11472906 DOI: 10.1161/circulationaha.123.067479] [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: 10/20/2023] [Accepted: 07/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The myocardium adapts to ischemia/reperfusion (I/R) by changes in gene expression, determining the cardiac response to reperfusion. mRNA translation is a key component of gene expression. It is largely unknown how regulation of mRNA translation contributes to cardiac gene expression and inflammation in response to reperfusion and whether it can be targeted to mitigate I/R injury. METHODS To examine translation and its impact on gene expression in response to I/R, we measured protein synthesis after reperfusion in vitro and in vivo. Underlying mechanisms of translational control were examined by pharmacological and genetic targeting of translation initiation in mice. Cell type-specific ribosome profiling was performed in mice that had been subjected to I/R to determine the impact of mRNA translation on the regulation of gene expression in cardiomyocytes. Translational regulation of inflammation was studied by quantification of immune cell infiltration, inflammatory gene expression, and cardiac function after short-term inhibition of translation initiation. RESULTS Reperfusion induced a rapid recovery of translational activity that exceeds baseline levels in the infarct and border zone and is mediated by translation initiation through the mTORC1 (mechanistic target of rapamycin complex 1)-4EBP1 (eIF4E-binding protein 1)-eIF (eukaryotic initiation factor) 4F axis. Cardiomyocyte-specific ribosome profiling identified that I/R increased translation of mRNA networks associated with cardiac inflammation and cell infiltration. Short-term inhibition of the mTORC1-4EBP1-eIF4F axis decreased the expression of proinflammatory cytokines such as Ccl2 (C-C motif chemokine ligand 2) of border zone cardiomyocytes, thereby attenuating Ly6Chi monocyte infiltration and myocardial inflammation. In addition, we identified a systemic immunosuppressive effect of eIF4F translation inhibitors on circulating monocytes, directly inhibiting monocyte infiltration. Short-term pharmacological inhibition of eIF4F complex formation by 4EGI-1 or rapamycin attenuated translation, reduced infarct size, and improved cardiac function after myocardial infarction. CONCLUSIONS Global protein synthesis is inhibited during ischemia and shortly after reperfusion, followed by a recovery of protein synthesis that exceeds baseline levels in the border and infarct zones. Activation of mRNA translation after reperfusion is driven by mTORC1/eIF4F-mediated regulation of initiation and mediates an mRNA network that controls inflammation and monocyte infiltration to the myocardium. Transient inhibition of the mTORC1-/eIF4F axis inhibits translation and attenuates Ly6Chi monocyte infiltration by inhibiting a proinflammatory response at the site of injury and of circulating monocytes.
Collapse
Affiliation(s)
- Christoph Hofmann
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
- Faculty of Biosciences, Heidelberg University, Germany (C.H.)
| | - Adrian Serafin
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
| | - Ole M. Schwerdt
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
| | - Johannes Fischer
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
| | - Florian Sicklinger
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
| | - Fereshteh S. Younesi
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
| | - Nikole J. Byrne
- University Heart Center Graz, Department of Cardiology, Medical University of Graz, Austria (N.J.B., H.B.)
| | - Ingmar S. Meyer
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
| | - Ellen Malovrh
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
| | - Clara Sandmann
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
| | - Lonny Jürgensen
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
| | - Verena Kamuf-Schenk
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
| | - Claudia Stroh
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
| | - Zoe Löwenthal
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
| | - Daniel Finke
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
| | - Etienne Boileau
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
| | - Arica Beisaw
- Department of Internal Medicine VIII (A.B.), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
| | - Heiko Bugger
- University Heart Center Graz, Department of Cardiology, Medical University of Graz, Austria (N.J.B., H.B.)
| | - Mandy Rettel
- Proteomics Core Facility, European Molecular Biology Laboratory Heidelberg, Germany (M.R., F. Stein)
| | - Frank Stein
- Proteomics Core Facility, European Molecular Biology Laboratory Heidelberg, Germany (M.R., F. Stein)
| | - Hugo A. Katus
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
| | - Tobias Jakobi
- Department of Internal Medicine and the Translational Cardiovascular Research Center, University of Arizona, College of Medicine–Phoenix (T.J.)
| | - Norbert Frey
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
| | - Florian Leuschner
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
| | - Mirko Völkers
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology [C.H., A.S., O.M.S., J.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., H.A.K., N.F., F.L., M.V.]), Heidelberg University Hospital, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (C.H., A.S., O.M.S., A.F., F. Sicklinger, F.S.Y., I.S.M., E.M., C. Sandmann, L.J., V.K.-S., C. Stroh, Z.L., D.F., E.B., A.B., H.A.K., N.F., F.L., M.V.)
| |
Collapse
|
13
|
Bobrovskikh AV, Zubairova US, Naumenko LG, Doroshkov AV. Catching the Big Fish in Big Data: A Meta-Analysis of Zebrafish Kidney scRNA-Seq Datasets Highlights Conserved Molecular Profiles of Macrophages and Neutrophils in Vertebrates. BIOLOGY 2024; 13:773. [PMID: 39452082 PMCID: PMC11505477 DOI: 10.3390/biology13100773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/10/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024]
Abstract
The innate immune system (IIS) is an ancient and essential defense mechanism that protects animals against a wide range of pathogens and diseases. Although extensively studied in mammals, our understanding of the IIS in other taxa remains limited. The zebrafish (Danio rerio) serves as a promising model organism for investigating IIS-related processes, yet the immunogenetics of fish are not fully elucidated. To address this gap, we conducted a meta-analysis of single-cell RNA sequencing (scRNA-seq) datasets from zebrafish kidney marrow, encompassing approximately 250,000 immune cells. Our analysis confirms the presence of key genetic pathways in zebrafish innate immune cells that are similar to those identified in mammals. Zebrafish macrophages specifically express genes encoding cathepsins, major histocompatibility complex class II proteins, integral membrane proteins, and the V-ATPase complex and demonstrate the enrichment of oxidative phosphorylation ferroptosis processes. Neutrophils are characterized by the significant expression of genes encoding actins, cytoskeleton organizing proteins, the Arp2/3 complex, and glycolysis enzymes and have demonstrated their involvement in GnRH and CLR signaling pathways, adherents, and tight junctions. Both macrophages and neutrophils highly express genes of NOD-like receptors, phagosomes, and lysosome pathways and genes involved in apoptosis. Our findings reinforce the idea about the existence of a wide spectrum of immune cell phenotypes in fish since we found only a small number of cells with clear pro- or anti-inflammatory signatures.
Collapse
Affiliation(s)
- Aleksandr V. Bobrovskikh
- Department of Physics, Novosibirsk State University, 630090 Novosibirsk, Russia;
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (U.S.Z.); (A.V.D.)
| | - Ulyana S. Zubairova
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (U.S.Z.); (A.V.D.)
- Department of Information Technologies, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Ludmila G. Naumenko
- Department of Physics, Novosibirsk State University, 630090 Novosibirsk, Russia;
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (U.S.Z.); (A.V.D.)
| | - Alexey V. Doroshkov
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (U.S.Z.); (A.V.D.)
- Department of Genomics and Bioinformatics, Institute of Fundamental Biology and Biotechnology, Siberian Federal University, 660036 Krasnoyarsk, Russia
| |
Collapse
|
14
|
Goumenaki P, Günther S, Kikhi K, Looso M, Marín-Juez R, Stainier DYR. The innate immune regulator MyD88 dampens fibrosis during zebrafish heart regeneration. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1158-1176. [PMID: 39271818 PMCID: PMC11399109 DOI: 10.1038/s44161-024-00538-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 08/06/2024] [Indexed: 09/15/2024]
Abstract
The innate immune response is triggered rapidly after injury and its spatiotemporal dynamics are critical for regeneration; however, many questions remain about its exact role. Here we show that MyD88, a key component of the innate immune response, controls not only the inflammatory but also the fibrotic response during zebrafish cardiac regeneration. We find in cryoinjured myd88-/- ventricles a significant reduction in neutrophil and macrophage numbers and the expansion of a collagen-rich endocardial population. Further analyses reveal compromised PI3K/AKT pathway activation in the myd88-/- endocardium and increased myofibroblasts and scarring. Notably, endothelial-specific overexpression of myd88 reverses these neutrophil, fibrotic and scarring phenotypes. Mechanistically, we identify the endocardial-derived chemokine gene cxcl18b as a target of the MyD88 signaling pathway, and using loss-of-function and gain-of-function tools, we show that it controls neutrophil recruitment. Altogether, these findings shed light on the pivotal role of MyD88 in modulating inflammation and fibrosis during tissue regeneration.
Collapse
Affiliation(s)
- Pinelopi Goumenaki
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- DZHK German Centre for Cardiovascular Research, Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Stefan Günther
- DZHK German Centre for Cardiovascular Research, Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Khrievono Kikhi
- Flow Cytometry Service Group, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Mario Looso
- DZHK German Centre for Cardiovascular Research, Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
- Bioinformatics Core Unit (BCU), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Rubén Marín-Juez
- Centre Hospitalier Universitaire Sainte-Justine Research Centre, Montreal, Quebec, Canada
- Department of Pathology and Cell Biology, University of Montreal, Montreal, Quebec, Canada
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
- DZHK German Centre for Cardiovascular Research, Partner Site Rhine-Main, Bad Nauheim, Germany.
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.
| |
Collapse
|
15
|
Lin L, Chu J, An S, Liu X, Tan R. The Biological Mechanisms and Clinical Roles of RNA-Binding Proteins in Cardiovascular Diseases. Biomolecules 2024; 14:1056. [PMID: 39334823 PMCID: PMC11430443 DOI: 10.3390/biom14091056] [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: 07/14/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
RNA-binding proteins (RBPs) have pivotal roles in cardiovascular biology, influencing various molecular mechanisms underlying cardiovascular diseases (CVDs). This review explores the significant roles of RBPs, focusing on their regulation of RNA alternative splicing, polyadenylation, and RNA editing, and their impact on CVD pathogenesis. For instance, RBPs are crucial in myocardial injury, contributing to disease progression and repair mechanisms. This review systematically analyzes the roles of RBPs in myocardial injury, arrhythmias, myocardial infarction, and heart failure, revealing intricate interactions that influence disease outcomes. Furthermore, the potential of RBPs as therapeutic targets for cardiovascular dysfunction is explored, highlighting the advances in drug development and clinical research. This review also discusses the emerging role of RBPs as biomarkers for cardiovascular diseases, offering insights into their diagnostic and prognostic potential. Despite significant progress, current research faces several limitations, which are critically examined. Finally, this review identifies the major challenges and outlines future research directions to advance the understanding and application of RBPs in cardiovascular medicine.
Collapse
Affiliation(s)
- Lizhu Lin
- Department of Anaesthesiology, The First People’s Hospital of Qinzhou, The Tenth Affiliated Hospital of Guangxi Medical University, Qinzhou 535000, China;
| | - Jiemei Chu
- Life Sciences Institute, Guangxi Medical University, Nanning 530021, China; (J.C.); (S.A.)
| | - Sanqi An
- Life Sciences Institute, Guangxi Medical University, Nanning 530021, China; (J.C.); (S.A.)
| | - Xinli Liu
- Life Sciences Institute, Guangxi Medical University, Nanning 530021, China; (J.C.); (S.A.)
| | - Runxian Tan
- Department of Laboratory Medicine, The First People’s Hospital of Qinzhou, The Tenth Affiliated Hospital of Guangxi Medical University, Qinzhou 535000, China
| |
Collapse
|
16
|
Kawaguchi A, Wang J, Knapp D, Murawala P, Nowoshilow S, Masselink W, Taniguchi-Sugiura Y, Fei JF, Tanaka EM. A chromatin code for limb segment identity in axolotl limb regeneration. Dev Cell 2024; 59:2239-2253.e9. [PMID: 38788714 DOI: 10.1016/j.devcel.2024.05.002] [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: 01/15/2023] [Revised: 07/25/2023] [Accepted: 05/03/2024] [Indexed: 05/26/2024]
Abstract
The salamander limb correctly regenerates missing limb segments because connective tissue cells have segment-specific identities, termed "positional information". How positional information is molecularly encoded at the chromatin level has been unknown. Here, we performed genome-wide chromatin profiling in mature and regenerating axolotl limb connective tissue cells. We find segment-specific levels of histone H3K27me3 as the major positional mark, especially at limb homeoprotein gene loci but not their upstream regulators, constituting an intrinsic segment information code. During regeneration, regeneration-specific regulatory elements became active prior to the re-appearance of developmental regulatory elements. In the hand, the permissive chromatin state of the homeoprotein gene HoxA13 engages with the regeneration program bypassing the upper limb program. Comparison of regeneration regulatory elements with those found in other regenerative animals identified a core shared set of transcription factors, supporting an ancient, conserved regeneration program.
Collapse
Affiliation(s)
- Akane Kawaguchi
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Jingkui Wang
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Dunja Knapp
- DFG Research Center for Regenerative Therapies, Technische Universität Dresden, 01307 Dresden, Germany
| | - Prayag Murawala
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria; DFG Research Center for Regenerative Therapies, Technische Universität Dresden, 01307 Dresden, Germany
| | - Sergej Nowoshilow
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria; DFG Research Center for Regenerative Therapies, Technische Universität Dresden, 01307 Dresden, Germany
| | - Wouter Masselink
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Yuka Taniguchi-Sugiura
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Ji-Feng Fei
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Elly M Tanaka
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), 1030 Vienna, Austria.
| |
Collapse
|
17
|
Huang K, Huang D, Li Q, Zeng J, Qin T, Zhong J, Zhong Z, Lu S. CircSorbs1 regulates myocardial regeneration and reduces cancer therapy-related cardiovascular toxicity through the Mir-99/GATA4 pathway. Discov Oncol 2024; 15:319. [PMID: 39080192 PMCID: PMC11289204 DOI: 10.1007/s12672-024-01075-0] [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: 04/06/2024] [Accepted: 06/03/2024] [Indexed: 08/02/2024] Open
Abstract
Due to the cancer therapy-related cardiovascular toxicity, heart failure following cancer therapy has a significant mortality rate. Gene-targeted therapy promotes the re-entry of existing cardiomyocytes into the cell cycle to achieve myocardial regeneration, which is a promising strategy for preventing and treating heart failure after myocardial infarction. Circular RNAs (circRNAs) are considered as potential targets for myocardial regeneration due to their strong stability, resistance to degradation, and potential role in heart development and cardiovascular diseases. By comparing the myocardial tissue of mice in the sham operation group and the Doxorubicin therapy group (DOX), we observed a significant decrease in Cirsorbs expression in the DOX group. Cirsorbs was predominantly localized in cardiomyocytes and exhibited high conservation. Subsequent investigations revealed that Cirsorbs could promote myocardial proliferation and inhibit myocardial apoptosis. Mechanistic studies further demonstrated that Cirsorbs could bind to miR99 and reduce its expression level. Meanwhile, miR99 was found to bind to GATA4 mRNA and decrease its expression level. The binding of Cirsorbs to miR99 alleviated the repression of miR99, thereby enhancing GATA4 expression and the transcription of downstream cyclin A2 and cyclin E1. This, in turn, increased cardiomyocyte proliferation and reduced apoptosis. In conclusion, Cirsorbs holds promise as an effective target for myocardial regeneration in reducing cancer therapy-related cardiovascular toxicity.
Collapse
Affiliation(s)
- Kang Huang
- Department of Cardiology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, No.43, Renmin Avenue, Haikou, 570208, Hainan, China
| | - Denggao Huang
- Central Laboratory, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, 570208, Hainan, China
| | - Qiang Li
- Department of Cardiology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, No.43, Renmin Avenue, Haikou, 570208, Hainan, China
| | - Jiangting Zeng
- Department of Cardiology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, No.43, Renmin Avenue, Haikou, 570208, Hainan, China
| | - Ting Qin
- Department of Cardiology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, No.43, Renmin Avenue, Haikou, 570208, Hainan, China
| | - Jianghua Zhong
- Department of Cardiology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, No.43, Renmin Avenue, Haikou, 570208, Hainan, China
| | - Zanrui Zhong
- Department of Cardiology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, No.43, Renmin Avenue, Haikou, 570208, Hainan, China
| | - Shijuan Lu
- Department of Cardiology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, No.43, Renmin Avenue, Haikou, 570208, Hainan, China.
| |
Collapse
|
18
|
Deng J, Yang JC, Feng Y, Xu ZJ, Kuča K, Liu M, Sun LH. AP-1 and SP1 trans-activate the expression of hepatic CYP1A1 and CYP2A6 in the bioactivation of AFB 1 in chicken. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1468-1478. [PMID: 38703348 DOI: 10.1007/s11427-023-2512-6] [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: 11/28/2023] [Accepted: 01/09/2024] [Indexed: 05/06/2024]
Abstract
Dietary exposure to aflatoxin B1 (AFB1) is harmful to the health and performance of domestic animals. The hepatic cytochrome P450s (CYPs), CYP1A1 and CYP2A6, are the primary enzymes responsible for the bioactivation of AFB1 to the highly toxic exo-AFB1-8,9-epoxide (AFBO) in chicks. However, the transcriptional regulation mechanism of these CYP genes in the liver of chicks in AFB1 metabolism remains unknown. Dual-luciferase reporter assay, bioinformatics and site-directed mutation results indicated that specificity protein 1 (SP1) and activator protein-1 (AP-1) motifs were located in the core region -1,063/-948, -606/-541 of the CYP1A1 promoter as well as -636/-595, -503/-462, -147/-1 of the CYP2A6 promoter. Furthermore, overexpression and decoy oligodeoxynucleotide technologies demonstrated that SP1 and AP-1 were pivotal transcriptional activators regulating the promoter activity of CYP1A1 and CYP2A6. Moreover, bioactivation of AFB1 to AFBO could be increased by upregulation of CYP1A1 and CYP2A6 expression, which was trans-activated owing to the upregulalion of AP-1, rather than SP1, stimulated by AFB1-induced reactive oxygen species. Additionally, nano-selenium could reduce ROS, downregulate AP-1 expression and then decrease the expression of CYP1A1 and CYP2A6, thus alleviating the toxicity of AFB1. In conclusion, AP-1 and SP1 played important roles in the transactivation of CYP1A1 and CYP2A6 expression and further bioactivated AFB1 to AFBO in chicken liver, which could provide novel targets for the remediation of aflatoxicosis in chicks.
Collapse
Affiliation(s)
- Jiang Deng
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jia-Cheng Yang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yue Feng
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ze-Jing Xu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Kamil Kuča
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
| | - Meng Liu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Lv-Hui Sun
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
19
|
Xiao Y, Hu L, Duan J, Che H, Wang W, Yuan Y, Xu J, Chen D, Zhao S. Polystyrene microplastics enhance microcystin-LR-induced cardiovascular toxicity and oxidative stress in zebrafish embryos. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 352:124022. [PMID: 38679130 DOI: 10.1016/j.envpol.2024.124022] [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: 01/28/2024] [Revised: 04/11/2024] [Accepted: 04/20/2024] [Indexed: 05/01/2024]
Abstract
The health risks associated with combined exposure to microplastics (MPs) and cyanobacteria toxins have gained increasing attention due to the large-scale prevalence of cyanobacterial blooms and accumulation of MPs in aquatic environments. Therefore, we explored the cardiovascular toxic effects of microcystin-LR (MC-LR, 1, 10, 100 μg/L) in the presence of 5 μm polystyrene microplastics (PS-MPs, 100 μg/L) and 80 nm polystyrene nanoplastics (PS-NPs, 100 μg/L) in zebrafish models. Embryos were exposed to certain PS-MPs and PS-NPs conditions in water between 3 h post-fertilization (hpf) and 168 hpf. Compared to MC-LR alone, a significant decrease in heart rate was observed as well as notable pericardial edema in the MC-LR + PS-MPs/NPs groups. At the same time, sinus venosus and bulbus arteriosus (SV-BA) distances were significantly increased. Furthermore, the addition of PS-MPs/NPs caused thrombosis in the caudal vein and more severe vascular damage in zebrafish larvae compared to MC-LR alone. Our findings revealed that combined exposure to PS-NPs and MC-LR could significantly decreased the expression of genes associated with cardiovascular development (myh6, nkx2.5, tnnt2a, and vegfaa), ATPase (atp1a3b, atp1b2b, atp2a1l, atp2b1a, and atp2b4), and the calcium channel (cacna1ab and ryr2a) compared to exposure to MC-LR alone. In addition, co-exposure with PS-MPs/NPs exacerbated the MC-LR-induced reactive oxygen species (ROS) production, as well as the ROS-stimulated apoptosis and heightened inflammation. We also discovered that astaxanthin (ASTA) treatment partially attenuated these cardiovascular toxic effects. Our findings confirm that exposure to MC-LR and PS-MPs/NPs affects cardiovascular development through calcium signaling interference and ROS-induced cardiovascular cell apoptosis. This study highlights the potential environmental risks of the co-existence of MC-LR and PS-MPs/NPs for fetal health, particularly cardiovascular development.
Collapse
Affiliation(s)
- Yuchun Xiao
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Liwen Hu
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Jiayao Duan
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Huimin Che
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Wenxin Wang
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Yuan Yuan
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Jiayi Xu
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Daojun Chen
- School of Medical Technology, Anhui Medical College, Hefei, 230601, China
| | - Sujuan Zhao
- School of Public Health, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
20
|
Shin K, Rodriguez-Parks A, Kim C, Silaban IM, Xia Y, Sun J, Dong C, Keles S, Wang J, Cao J, Kang J. Harnessing the regenerative potential of interleukin11 to enhance heart repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.29.577788. [PMID: 38352555 PMCID: PMC10862709 DOI: 10.1101/2024.01.29.577788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Balancing between regenerative processes and fibrosis is crucial for heart repair, yet strategies regulating this balance remain a barrier to developing therapies. While Interleukin11 (IL11) is known as a fibrotic factor, its contribution to heart regeneration is poorly understood. We uncovered that il11a, an Il11 homolog in zebrafish, can trigger robust regenerative programs in zebrafish hearts, including cardiomyocytes proliferation and coronary expansion, even in the absence of injury. However, prolonged il11a induction in uninjured hearts causes persistent fibroblast emergence, resulting in fibrosis. While deciphering the regenerative and fibrotic effects of il11a, we found that il11-dependent fibrosis, but not regeneration, is mediated through ERK activity, suggesting to potentially uncouple il11a dual effects on regeneration and fibrosis. To harness the il11a's regenerative ability, we devised a combinatorial treatment through il11a induction with ERK inhibition. This approach enhances cardiomyocyte proliferation with mitigated fibrosis, achieving a balance between regenerative processes and fibrosis. Thus, we unveil the mechanistic insights into regenerative il11 roles, offering therapeutic avenues to foster cardiac repair without exacerbating fibrosis.
Collapse
Affiliation(s)
- Kwangdeok Shin
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, 53705, USA
| | - Anjelica Rodriguez-Parks
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, 53705, USA
| | - Chanul Kim
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, 53705, USA
| | - Isabella M Silaban
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, 53705, USA
| | - Yu Xia
- Cardiovascular Research Institute, Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Jisheng Sun
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Chenyang Dong
- Departments of Statistics and of Biostatistics and Medical Informatics, University of Wisconsin - Madison, Madison, WI 53706, USA
| | - Sunduz Keles
- Departments of Statistics and of Biostatistics and Medical Informatics, University of Wisconsin - Madison, Madison, WI 53706, USA
| | - Jinhu Wang
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Jingli Cao
- Cardiovascular Research Institute, Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Junsu Kang
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, 53705, USA
- UW Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, 53705, USA
| |
Collapse
|
21
|
Gao X, Xuan Y, Zhou Z, Chen C, Wen Wang D, Wen Z. Ivermectin ameliorates acute myocarditis via the inhibition of importin-mediated nuclear translocation of NF-κB/p65. Int Immunopharmacol 2024; 133:112073. [PMID: 38636372 DOI: 10.1016/j.intimp.2024.112073] [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: 12/09/2023] [Revised: 03/24/2024] [Accepted: 04/09/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Myocarditis is an important clinical issue which lacks specific treatment by now. Ivermectin (IVM) is an inhibitor of importin α/β-mediated nuclear translocation. This study aimed to explore the therapeutic effects of IVM on acute myocarditis. METHODS Mouse models of coxsackie B3 virus (CVB3) infection-induced myocarditis and experimental autoimmune myocarditis (EAM) were established to evaluate the effects of IVM. Cardiac functions were evaluated by echocardiography and Millar catheter. Cardiac inflammatory infiltration was assessed by histological staining. Cytometric bead array and quantitative real-time PCR were used to detect the levels of pro-inflammatory cytokines. The macrophages and their M1/M2 polarization were analyzed via flow cytometry. Protein expression and binding were detected by co-immunoprecipitation, Western blotting and histological staining. The underlying mechanism was verified in vitro using CVB3-infected RAW264.7 macrophages. Cyclic polypeptide (cTN50) was synthesized to selectively inhibit the nuclear translocation of NF-κB/p65, and CVB3-infected RAW264.7 cells were treated with cTN50. RESULTS Increased expression of importin β was observed in both models. IVM treatment improved cardiac functions and reduced the cardiac inflammation associated with CVB3-myocarditis and EAM. Furthermore, the pro-inflammatory cytokine (IL-1β/IL-6/TNF-α) levels were downregulated via the inhibition of the nuclear translocation of NF-κB/p65 in macrophages. IVM and cTN50 treatment also inhibited the nuclear translocation of NF-κB/p65 and downregulated the expression of pro-inflammatory cytokines in RAW264.7 macrophages. CONCLUSIONS Ivermectin inhibits the nuclear translocation of NF-κB/p65 and the expression of major pro-inflammatory cytokines in myocarditis. The therapeutic effects of IVM on viral and non-viral myocarditis models suggest its potential application in the treatment of acute myocarditis.
Collapse
Affiliation(s)
- Xu Gao
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, PR China
| | - Yunling Xuan
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, PR China
| | - Zhou Zhou
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, PR China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, PR China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, PR China
| | - Zheng Wen
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, PR China.
| |
Collapse
|
22
|
Cordero J, Elsherbiny A, Wang Y, Jürgensen L, Constanty F, Günther S, Boerries M, Heineke J, Beisaw A, Leuschner F, Hassel D, Dobreva G. Leveraging chromatin state transitions for the identification of regulatory networks orchestrating heart regeneration. Nucleic Acids Res 2024; 52:4215-4233. [PMID: 38364861 PMCID: PMC11077086 DOI: 10.1093/nar/gkae085] [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: 05/26/2023] [Revised: 01/23/2024] [Accepted: 02/06/2024] [Indexed: 02/18/2024] Open
Abstract
The limited regenerative capacity of the human heart contributes to high morbidity and mortality worldwide. In contrast, zebrafish exhibit robust regenerative capacity, providing a powerful model for studying how to overcome intrinsic epigenetic barriers maintaining cardiac homeostasis and initiate regeneration. Here, we present a comprehensive analysis of the histone modifications H3K4me1, H3K4me3, H3K27me3 and H3K27ac during various stages of zebrafish heart regeneration. We found a vast gain of repressive chromatin marks one day after myocardial injury, followed by the acquisition of active chromatin characteristics on day four and a transition to a repressive state on day 14, and identified distinct transcription factor ensembles associated with these events. The rapid transcriptional response involves the engagement of super-enhancers at genes implicated in extracellular matrix reorganization and TOR signaling, while H3K4me3 breadth highly correlates with transcriptional activity and dynamic changes at genes involved in proteolysis, cell cycle activity, and cell differentiation. Using loss- and gain-of-function approaches, we identified transcription factors in cardiomyocytes and endothelial cells influencing cardiomyocyte dedifferentiation or proliferation. Finally, we detected significant evolutionary conservation between regulatory regions that drive zebrafish and neonatal mouse heart regeneration, suggesting that reactivating transcriptional and epigenetic networks converging on these regulatory elements might unlock the regenerative potential of adult human hearts.
Collapse
Affiliation(s)
- Julio Cordero
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Adel Elsherbiny
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yinuo Wang
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lonny Jürgensen
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Florian Constanty
- Institute of Experimental Cardiology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Stefan Günther
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
- German Cancer Consortium (DKTK), Partner site Freiburg, a partnership between DKFZ and Medical Center - University of Freiburg, 69110 Heidelberg, Germany
| | - Joerg Heineke
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Arica Beisaw
- Institute of Experimental Cardiology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Florian Leuschner
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - David Hassel
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Gergana Dobreva
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| |
Collapse
|
23
|
Jia JY, Chen GH, Shu TT, Lou QY, Jin X, He JY, Xiao WH, Zhai G, Yin Z. Androgen signaling inhibits de novo lipogenesis to alleviate lipid deposition in zebrafish. Zool Res 2024; 45:355-366. [PMID: 38485505 PMCID: PMC11017085 DOI: 10.24272/j.issn.2095-8137.2023.324] [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: 11/28/2023] [Accepted: 01/15/2024] [Indexed: 03/19/2024] Open
Abstract
Testosterone is closely associated with lipid metabolism and known to affect body fat composition and muscle mass in males. However, the mechanisms by which testosterone acts on lipid metabolism are not yet fully understood, especially in teleosts. In this study, cyp17a1-/- zebrafish ( Danio rerio) exhibited excessive visceral adipose tissue (VAT), lipid content, and up-regulated expression and activity of hepatic de novo lipogenesis (DNL) enzymes. The assay for transposase accessible chromatin with sequencing (ATAC-seq) results demonstrated that chromatin accessibility of DNL genes was increased in cyp17a1-/- fish compared to cyp17a1+/+ male fish, including stearoyl-CoA desaturase ( scd) and fatty acid synthase ( fasn). Androgen response element (ARE) motifs in the androgen signaling pathway were significantly enriched in cyp17a1+/+ male fish but not in cyp17a1-/- fish. Both androgen receptor ( ar)-/- and wild-type (WT) zebrafish administered with Ar antagonist flutamide displayed excessive visceral adipose tissue, lipid content, and up-regulated expression and activity of hepatic de novo lipogenesis enzymes. The Ar agonist BMS-564929 reduced the content of VAT and lipid content, and down-regulated acetyl-CoA carboxylase a ( acaca), fasn, and scd expression. Mechanistically, the rescue effect of testosterone on cyp17a1-/- fish in terms of phenotypes was abolished when ar was additionally depleted. Collectively, these findings reveal that testosterone inhibits lipid deposition by down-regulating DNL genes via Ar in zebrafish, thus expanding our understanding of the relationship between testosterone and lipid metabolism in teleosts.
Collapse
Affiliation(s)
- Jing-Yi Jia
- College of Fisheries, Huazhong Agriculture University, Wuhan, Hubei 430070, China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072, China
| | - Guang-Hui Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072, China
| | - Ting-Ting Shu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Hubei Key Laboratory of Three Gorges Project for Conservation of Fishes, Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, Hubei 443100, China
| | - Qi-Yong Lou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072, China
| | - Xia Jin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072, China
| | - Jiang-Yan He
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072, China
| | - Wu-Han Xiao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072, China
| | - Gang Zhai
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Hubei Hongshan Laboratory, Huazhong Agriculture University, Wuhan, Hubei 430070, China. E-mail:
| | - Zhan Yin
- College of Fisheries, Huazhong Agriculture University, Wuhan, Hubei 430070, China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Hubei Hongshan Laboratory, Huazhong Agriculture University, Wuhan, Hubei 430070, China
- Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing 100049, China. E-mail:
| |
Collapse
|
24
|
Constanty F, Wu B, Wei KH, Lin IT, Dallmann J, Guenther S, Lautenschlaeger T, Priya R, Lai SL, Stainier DYR, Beisaw A. Border-zone cardiomyocytes and macrophages contribute to remodeling of the extracellular matrix to promote cardiomyocyte invasion during zebrafish cardiac regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584570. [PMID: 38559277 PMCID: PMC10980021 DOI: 10.1101/2024.03.12.584570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Despite numerous advances in our understanding of zebrafish cardiac regeneration, an aspect that remains less studied is how regenerating cardiomyocytes invade, and eventually replace, the collagen-containing fibrotic tissue following injury. Here, we provide an in-depth analysis of the process of cardiomyocyte invasion using live-imaging and histological approaches. We observed close interactions between protruding cardiomyocytes and macrophages at the wound border zone, and macrophage-deficient irf8 mutant zebrafish exhibited defects in extracellular matrix (ECM) remodeling and cardiomyocyte protrusion into the injured area. Using a resident macrophage ablation model, we show that defects in ECM remodeling at the border zone and subsequent cardiomyocyte protrusion can be partly attributed to a population of resident macrophages. Single-cell RNA-sequencing analysis of cells at the wound border revealed a population of cardiomyocytes and macrophages with fibroblast-like gene expression signatures, including the expression of genes encoding ECM structural proteins and ECM-remodeling proteins. The expression of mmp14b , which encodes a membrane-anchored matrix metalloproteinase, was restricted to cells in the border zone, including cardiomyocytes, macrophages, fibroblasts, and endocardial/endothelial cells. Genetic deletion of mmp14b led to a decrease in 1) macrophage recruitment to the border zone, 2) collagen degradation at the border zone, and 3) subsequent cardiomyocyte invasion. Furthermore, cardiomyocyte-specific overexpression of mmp14b was sufficient to enhance cardiomyocyte invasion into the injured tissue and along the apical surface of the wound. Altogether, our data shed important insights into the process of cardiomyocyte invasion of the collagen-containing injured tissue during cardiac regeneration. They further suggest that cardiomyocytes and resident macrophages contribute to ECM remodeling at the border zone to promote cardiomyocyte replenishment of the fibrotic injured tissue.
Collapse
|
25
|
Ando K, Ou J, Thompson JD, Welsby J, Bangru S, Shen J, Wei X, Diao Y, Poss KD. A screen for regeneration-associated silencer regulatory elements in zebrafish. Dev Cell 2024; 59:676-691.e5. [PMID: 38290519 PMCID: PMC10939760 DOI: 10.1016/j.devcel.2024.01.004] [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: 06/24/2023] [Revised: 11/03/2023] [Accepted: 01/08/2024] [Indexed: 02/01/2024]
Abstract
Regeneration involves gene expression changes explained in part by context-dependent recruitment of transcriptional activators to distal enhancers. Silencers that engage repressive transcriptional complexes are less studied than enhancers and more technically challenging to validate, but they potentially have profound biological importance for regeneration. Here, we identified candidate silencers through a screening process that examined the ability of DNA sequences to limit injury-induced gene expression in larval zebrafish after fin amputation. A short sequence (s1) on chromosome 5 near several genes that reduce expression during adult fin regeneration could suppress promoter activity in stable transgenic lines and diminish nearby gene expression in knockin lines. High-resolution analysis of chromatin organization identified physical associations of s1 with gene promoters occurring preferentially during fin regeneration, and genomic deletion of s1 elevated the expression of these genes after fin amputation. Our study provides methods to identify "tissue regeneration silencer elements" (TRSEs) with the potential to reduce unnecessary or deleterious gene expression during regeneration.
Collapse
Affiliation(s)
- Kazunori Ando
- Duke Regeneration Center and Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jianhong Ou
- Duke Regeneration Center and Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - John D Thompson
- Duke Regeneration Center and Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - John Welsby
- Duke Regeneration Center and Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sushant Bangru
- Duke Regeneration Center and Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jingwen Shen
- Duke Regeneration Center and Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Xiaolin Wei
- Duke Regeneration Center and Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yarui Diao
- Duke Regeneration Center and Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kenneth D Poss
- Duke Regeneration Center and Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
26
|
Smits AM, Bollini S, Gladka MM. Editorial: Novel strategies to repair the infarcted heart, volume II. Front Cardiovasc Med 2024; 11:1379240. [PMID: 38516001 PMCID: PMC10955122 DOI: 10.3389/fcvm.2024.1379240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 02/28/2024] [Indexed: 03/23/2024] Open
Affiliation(s)
- Anke M. Smits
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Sveva Bollini
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Monika M. Gladka
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| |
Collapse
|
27
|
Dong Y, Yang Y, Wang H, Feng D, Nist E, Yapundich N, Spurlock B, Craft M, Qian L, Liu J. Single-cell chromatin profiling reveals genetic programs activating proregenerative states in nonmyocyte cells. SCIENCE ADVANCES 2024; 10:eadk4694. [PMID: 38381829 PMCID: PMC10881044 DOI: 10.1126/sciadv.adk4694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 01/18/2024] [Indexed: 02/23/2024]
Abstract
Cardiac regeneration requires coordinated participation of multiple cell types whereby their communications result in transient activation of proregenerative cell states. Although the molecular characteristics and lineage origins of these activated cell states and their contribution to cardiac regeneration have been studied, the extracellular signaling and the intrinsic genetic program underlying the activation of the transient functional cell states remain largely unexplored. In this study, we delineated the chromatin landscapes of the noncardiomyocytes (nonCMs) of the regenerating heart at the single-cell level and inferred the cis-regulatory architectures and trans-acting factors that control cell type-specific gene expression programs. Moreover, further motif analysis and cell-specific genetic manipulations suggest that the macrophage-derived inflammatory signal tumor necrosis factor-α, acting via its downstream transcription factor complex activator protein-1, functions cooperatively with discrete transcription regulators to activate respective nonCM cell types critical for cardiac regeneration. Thus, our study defines the regulatory architectures and intercellular communication principles in zebrafish heart regeneration.
Collapse
Affiliation(s)
- Yanhan Dong
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yuchen Yang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Haofei Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Dong Feng
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Elizabeth Nist
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Nicholas Yapundich
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Brian Spurlock
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Madison Craft
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
28
|
Weinberger M, Simões FC, Gungoosingh T, Sauka-Spengler T, Riley PR. Distinct epicardial gene regulatory programs drive development and regeneration of the zebrafish heart. Dev Cell 2024; 59:351-367.e6. [PMID: 38237592 DOI: 10.1016/j.devcel.2023.12.012] [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: 08/05/2022] [Revised: 08/12/2023] [Accepted: 12/20/2023] [Indexed: 02/08/2024]
Abstract
Unlike the adult mammalian heart, which has limited regenerative capacity, the zebrafish heart fully regenerates following injury. Reactivation of cardiac developmental programs is considered key to successfully regenerating the heart, yet the regulation underlying the response to injury remains elusive. Here, we compared the transcriptome and epigenome of the developing and regenerating zebrafish epicardia. We identified epicardial enhancer elements with specific activity during development or during adult heart regeneration. By generating gene regulatory networks associated with epicardial development and regeneration, we inferred genetic programs driving each of these processes, which were largely distinct. Loss of Hif1ab, Nrf1, Tbx2b, and Zbtb7a, central regulators of the regenerating epicardial network, in injured hearts resulted in elevated epicardial cell numbers infiltrating the wound and excess fibrosis after cryoinjury. Our work identifies differences between the regulatory blueprint deployed during epicardial development and regeneration, underlining that heart regeneration goes beyond the reactivation of developmental programs.
Collapse
Affiliation(s)
- Michael Weinberger
- Department of Physiology, Anatomy and Genetics, Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford OX3 7TY, Oxfordshire, UK; Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, Oxfordshire, UK
| | - Filipa C Simões
- Department of Physiology, Anatomy and Genetics, Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford OX3 7TY, Oxfordshire, UK
| | - Trishalee Gungoosingh
- Department of Physiology, Anatomy and Genetics, Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford OX3 7TY, Oxfordshire, UK
| | - Tatjana Sauka-Spengler
- Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, Oxfordshire, UK; Stowers Institute for Medical Research, Kansas City, MO, USA.
| | - Paul R Riley
- Department of Physiology, Anatomy and Genetics, Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford OX3 7TY, Oxfordshire, UK.
| |
Collapse
|
29
|
Ren Y, Xu Y, Wang Z, Wang Y, Zhang J, Li Z, Chen Y, Go W, Javed MT, Li Q. Molecular cloning, biological description, and functional analysis of Ajfos transcription factor in pathogen-induced Apostichopus japonicus. Comp Biochem Physiol C Toxicol Pharmacol 2024; 276:109814. [PMID: 38065305 DOI: 10.1016/j.cbpc.2023.109814] [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: 10/10/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024]
Abstract
Activator protein-1 subfamily member c-Fos wields significant influence over cellular activities, such as regulation of cell growth and division, cell death, and immune responses under various extracellular situations. In this study, the full-length c-Fos of sea cucumber, Apostichopus japonicus (Ajfos) was successfully cloned and analyzed. The anticipated 306 amino acid sequences of Ajfos displayed a basic-leucine zipper (bZIP) domain, similar to invertebrate counterparts. In addition, the qPCR results suggested Ajfos expressed in all tissues, with the highest level in coelomocytes from polian vesicle (vesicle lumen cells), followed by coelomocytes from coelom (coelomocytes). Moreover, the expression levels of Ajfos in the coelomocytes and vesicle lumen cells of sea cucumber showed significant changes after the Vibrio splendidus challenge, especially reaching a peak at 6 h. Compared with the silencing negative control RNA interference (siNC) group, silencing Ajfos (siAjfos) in vivo decreased the downstream proliferation-related gene expression of vesicle lumen cells after infection with V. splendidus while no significant influence was observed on coelomocytes. Furthermore, the proliferation proportion of vesicle lumen cells in the siAjfos group was significantly reduced under pathogen stimulation conditions. Finally, based on the fluctuation trend of total coelomocyte density (TCD) from coelom and polian vesicle previously discovered, it is evident that Ajfos played a critical role in facilitating the swift proliferation of vesicle lumen cells in response to V. splendidus stimulation. Altogether, this research provided an initial reference of the function of Ajfos in echinoderms, unveiling its participation in host coelomocyte proliferation of sea cucumbers during bacterial challenges.
Collapse
Affiliation(s)
- Yuan Ren
- School of Bioengineering, Dalian University of Technology, Dalian 116024, PR China
| | - Yongping Xu
- School of Bioengineering, Dalian University of Technology, Dalian 116024, PR China
| | - Zhenhui Wang
- College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng 224051, PR China
| | - Yinan Wang
- College of Fisheries, Tianjin Agricultural University, Tianjin 300392, PR China; College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng 224051, PR China
| | - Jialin Zhang
- College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng 224051, PR China
| | - Zhen Li
- College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng 224051, PR China
| | - Yupeng Chen
- College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng 224051, PR China
| | - Wei Go
- College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng 224051, PR China
| | - Muhammad Tariq Javed
- Faculty of Veterinary Science, University of Agriculture, Faisalabad 38040, Pakistan
| | - Qiang Li
- College of Fisheries, Tianjin Agricultural University, Tianjin 300392, PR China; College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng 224051, PR China.
| |
Collapse
|
30
|
Zhang R, Li Y, Zhang J. Molecular mechanisms of pelvic organ prolapse influenced by FBLN5 via FOSL1/miR-222/MEIS1/COL3A1 axis. Cell Signal 2024; 114:111000. [PMID: 38056607 DOI: 10.1016/j.cellsig.2023.111000] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/27/2023] [Accepted: 12/01/2023] [Indexed: 12/08/2023]
Abstract
This study delves into the role of FBLN5 in pelvic organ prolapse (POP) and its molecular mechanisms, focusing on the FOSL1/miR-222/MEIS1/COL3A1 axis. Gene relationships linked to POP were confirmed using bioinformatics databases like GEO and StarBase. Primary human uterosacral ligament fibroblasts (hUSLF) were extracted and subjected to mechanical stretching. Cellular cytoskeletal changes were examined via phalloidin staining, intracellular ROS levels with a ROS kit, cell apoptosis through flow cytometry, and cell senescence using β-galactosidase staining. FBLN5's downstream targets were identified, and the interaction between FOSL1 and miR-222 and miR-222 and MEIS1 were validated using assays. In rat models, the role of FBLN5 in POP was assessed using bladder pressure tests. Results indicated diminished FBLN5 expression in uterine prolapse. Enhanced FBLN5 countered mechanical damage in hUSLF cells by downregulating FOSL1. FOSL1 augmented miR-222, inhibiting MEIS1, which subsequently fostered COL3A1 transcription. In rat models, the absence of FBLN5 exacerbated POP by influencing the FOSL1/miR-222/MEIS1/COL3A1 pathway. FBLN5's protective role likely involves regulating the above axis and boosting COL3A1 expression. Further research is needed to validate the effectiveness and safety of this mechanism in human patients and to propose potential new treatment options.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Obstetrics and Gynecology, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing 100038, PR China
| | - Ya Li
- Department of Obstetrics and Gynecology, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing 100038, PR China
| | - Jin Zhang
- Department of Obstetrics and Gynecology, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing 100038, PR China.
| |
Collapse
|
31
|
Rolland L, Abaroa JM, Faucherre A, Drouard A, Jopling C. The ion channel Trpc6a regulates the cardiomyocyte regenerative response to mechanical stretch. Front Cardiovasc Med 2024; 10:1186086. [PMID: 38259319 PMCID: PMC10801195 DOI: 10.3389/fcvm.2023.1186086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 10/26/2023] [Indexed: 01/24/2024] Open
Abstract
Myocardial damage caused, for example, by cardiac ischemia leads to ventricular volume overload resulting in increased stretch of the remaining myocardium. In adult mammals, these changes trigger an adaptive cardiomyocyte hypertrophic response which, if the damage is extensive, will ultimately lead to pathological hypertrophy and heart failure. Conversely, in response to extensive myocardial damage, cardiomyocytes in the adult zebrafish heart and neonatal mice proliferate and completely regenerate the damaged myocardium. We therefore hypothesized that in adult zebrafish, changes in mechanical loading due to myocardial damage may act as a trigger to induce cardiac regeneration. Based on this notion we sought to identify mechanosensors which could be involved in detecting changes in mechanical loading and triggering regeneration. Here we show using a combination of knockout animals, RNAseq and in vitro assays that the mechanosensitive ion channel Trpc6a is required by cardiomyocytes for successful cardiac regeneration in adult zebrafish. Furthermore, using a cyclic cell stretch assay, we have determined that Trpc6a induces the expression of components of the AP1 transcription complex in response to mechanical stretch. Our data highlights how changes in mechanical forces due to myocardial damage can be detected by mechanosensors which in turn can trigger cardiac regeneration.
Collapse
Affiliation(s)
| | | | | | | | - Chris Jopling
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, LabEx ICST, Montpellier, France
| |
Collapse
|
32
|
Shin K, Begeman IJ, Cao J, Kang J. leptin b and its regeneration enhancer illustrate the regenerative features of zebrafish hearts. Dev Dyn 2024; 253:91-106. [PMID: 36495292 PMCID: PMC10256838 DOI: 10.1002/dvdy.556] [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: 07/14/2022] [Revised: 11/11/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Zebrafish possess a remarkable regenerative capacity, which is mediated by the induction of various genes upon injury. Injury-dependent transcription is governed by the tissue regeneration enhancer elements (TREEs). Here, we utilized leptin b (lepb), an injury-specific factor, and its TREE to dissect heterogeneity of noncardiomyocytes (CMs) in regenerating hearts. RESULTS Our single-cell RNA sequencing (scRNA-seq) analysis demonstrated that the endothelium/endocardium(EC) is activated to induce distinct subpopulations upon injury. We demonstrated that lepb can be utilized as a regeneration-specific marker to subset injury-activated ECs. lepb+ ECs robustly induce pro-regenerative factors, implicating lepb+ ECs as a signaling center to interact with other cardiac cells. Our scRNA-seq analysis identified that lepb is also produced by subpopulation of epicardium (Epi) and epicardium-derived cells (EPDCs). To determine whether lepb labels injury-emerging non-CM cells, we tested the activity of lepb-linked regeneration enhancer (LEN) with chromatin accessibility profiles and transgenic lines. While nondetectable in uninjured hearts, LEN directs EC and Epi/EPDC expression upon injury. The endogenous LEN activity was assessed using LEN deletion lines, demonstrating that LEN deletion abolished injury-dependent expression of lepb, but not other nearby genes. CONCLUSIONS Our integrative analyses identify regeneration-emerging cell-types and factors, leading to the discovery of regenerative features of hearts.
Collapse
Affiliation(s)
- Kwangdeok Shin
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, 53705, USA
| | - Ian J. Begeman
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, 53705, USA
| | - Jingli Cao
- Cardiovascular Research Institute, Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10021, USA
| | - Junsu Kang
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, 53705, USA
| |
Collapse
|
33
|
Beisaw A, Wu CC. Cardiomyocyte maturation and its reversal during cardiac regeneration. Dev Dyn 2024; 253:8-27. [PMID: 36502296 DOI: 10.1002/dvdy.557] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 12/03/2022] [Accepted: 12/03/2022] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular disease is a leading cause of death worldwide. Due to the limited proliferative and regenerative capacity of adult cardiomyocytes, the lost myocardium is not replenished efficiently and is replaced by a fibrotic scar, which eventually leads to heart failure. Current therapies to cure or delay the progression of heart failure are limited; hence, there is a pressing need for regenerative approaches to support the failing heart. Cardiomyocytes undergo a series of transcriptional, structural, and metabolic changes after birth (collectively termed maturation), which is critical for their contractile function but limits the regenerative capacity of the heart. In regenerative organisms, cardiomyocytes revert from their terminally differentiated state into a less mature state (ie, dedifferentiation) to allow for proliferation and regeneration to occur. Importantly, stimulating adult cardiomyocyte dedifferentiation has been shown to promote morphological and functional improvement after myocardial infarction, further highlighting the importance of cardiomyocyte dedifferentiation in heart regeneration. Here, we review several hallmarks of cardiomyocyte maturation, and summarize how their reversal facilitates cardiomyocyte proliferation and heart regeneration. A detailed understanding of how cardiomyocyte dedifferentiation is regulated will provide insights into therapeutic options to promote cardiomyocyte de-maturation and proliferation, and ultimately heart regeneration in mammals.
Collapse
Affiliation(s)
- Arica Beisaw
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany
| | - Chi-Chung Wu
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
34
|
Ram A, Murphy D, DeCuzzi N, Patankar M, Hu J, Pargett M, Albeck JG. A guide to ERK dynamics, part 2: downstream decoding. Biochem J 2023; 480:1909-1928. [PMID: 38038975 PMCID: PMC10754290 DOI: 10.1042/bcj20230277] [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: 07/09/2023] [Revised: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 12/02/2023]
Abstract
Signaling by the extracellular signal-regulated kinase (ERK) pathway controls many cellular processes, including cell division, death, and differentiation. In this second installment of a two-part review, we address the question of how the ERK pathway exerts distinct and context-specific effects on multiple processes. We discuss how the dynamics of ERK activity induce selective changes in gene expression programs, with insights from both experiments and computational models. With a focus on single-cell biosensor-based studies, we summarize four major functional modes for ERK signaling in tissues: adjusting the size of cell populations, gradient-based patterning, wave propagation of morphological changes, and diversification of cellular gene expression states. These modes of operation are disrupted in cancer and other related diseases and represent potential targets for therapeutic intervention. By understanding the dynamic mechanisms involved in ERK signaling, there is potential for pharmacological strategies that not only simply inhibit ERK, but also restore functional activity patterns and improve disease outcomes.
Collapse
Affiliation(s)
- Abhineet Ram
- Department of Molecular and Cellular Biology, University of California, Davis, CA, U.S.A
| | - Devan Murphy
- Department of Molecular and Cellular Biology, University of California, Davis, CA, U.S.A
| | - Nicholaus DeCuzzi
- Department of Molecular and Cellular Biology, University of California, Davis, CA, U.S.A
| | - Madhura Patankar
- Department of Molecular and Cellular Biology, University of California, Davis, CA, U.S.A
| | - Jason Hu
- Department of Molecular and Cellular Biology, University of California, Davis, CA, U.S.A
| | - Michael Pargett
- Department of Molecular and Cellular Biology, University of California, Davis, CA, U.S.A
| | - John G. Albeck
- Department of Molecular and Cellular Biology, University of California, Davis, CA, U.S.A
| |
Collapse
|
35
|
Suzuki S, Tanaka S, Kametani Y, Umeda A, Nishinaka K, Egawa K, Okada Y, Obana M, Fujio Y. Runx1 is upregulated by STAT3 and promotes proliferation of neonatal rat cardiomyocytes. Physiol Rep 2023; 11:e15872. [PMID: 38040660 PMCID: PMC10691971 DOI: 10.14814/phy2.15872] [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: 08/14/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 12/03/2023] Open
Abstract
Though it is well known that mammalian cardiomyocytes exit cell cycle soon after birth, the mechanisms that regulate proliferation remain to be fully elucidated. Recent studies reported that cardiomyocytes undergo dedifferentiation before proliferation, indicating the importance of dedifferentiation in cardiomyocyte proliferation. Since Runx1 is expressed in dedifferentiated cardiomyocytes, Runx1 is widely used as a dedifferentiation marker of cardiomyocytes; however, little is known about the role of Runx1 in the proliferation of cardiomyocytes. The purpose of this study was to clarify the functional significance of Runx1 in cardiomyocyte proliferation. qRT-PCR analysis and immunoblot analysis demonstrated that Runx1 expression was upregulated in neonatal rat cardiomyocytes when cultured in the presence of FBS. Similarly, STAT3 was activated in the presence of FBS. Interestingly, knockdown of STAT3 significantly decreased Runx1 expression, indicating Runx1 is regulated by STAT3. We next investigated the effect of Runx1 on proliferation. Immunofluorescence microscopic analysis using an anti-Ki-67 antibody revealed that knockdown of Runx1 decreased the ratio of proliferating cardiomyocytes. Conversely, Runx1 overexpression using adenovirus vector induced cardiomyocyte proliferation in the absence of FBS. Finally, RNA-sequencing analysis revealed that Runx1 overexpression induced upregulation of cardiac fetal genes and downregulation of genes associated with fatty acid oxidation. Collectively, Runx1 is regulated by STAT3 and induces cardiomyocyte proliferation by juvenilizing cardiomyocytes.
Collapse
Affiliation(s)
- Shota Suzuki
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical SciencesOsaka UniversitySuita CityOsakaJapan
| | - Shota Tanaka
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical SciencesOsaka UniversitySuita CityOsakaJapan
| | - Yusuke Kametani
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical SciencesOsaka UniversitySuita CityOsakaJapan
| | - Ayaka Umeda
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical SciencesOsaka UniversitySuita CityOsakaJapan
| | - Kosuke Nishinaka
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical SciencesOsaka UniversitySuita CityOsakaJapan
| | - Kaho Egawa
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical SciencesOsaka UniversitySuita CityOsakaJapan
| | - Yoshiaki Okada
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical SciencesOsaka UniversitySuita CityOsakaJapan
- Center for Infectious Disease Education and Research (CiDER)Osaka UniversitySuita CityOsakaJapan
| | - Masanori Obana
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical SciencesOsaka UniversitySuita CityOsakaJapan
- Center for Infectious Disease Education and Research (CiDER)Osaka UniversitySuita CityOsakaJapan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI)Osaka UniversitySuita CityOsakaJapan
- Global Center for Medical Engineering and Informatics (MEI)Osaka UniversitySuita CityOsakaJapan
- Radioisotope Research Center, Institute for Radiation SciencesOsaka UniversitySuita CityOsakaJapan
| | - Yasushi Fujio
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical SciencesOsaka UniversitySuita CityOsakaJapan
- Center for Infectious Disease Education and Research (CiDER)Osaka UniversitySuita CityOsakaJapan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI)Osaka UniversitySuita CityOsakaJapan
| |
Collapse
|
36
|
Zhong H, Zhang R, Li G, Huang P, Zhang Y, Zhu J, Kuang J, Hutchins AP, Qin D, Zhu P, Pei D, Li D. c-JUN is a barrier in hESC to cardiomyocyte transition. Life Sci Alliance 2023; 6:e202302121. [PMID: 37604584 PMCID: PMC10442936 DOI: 10.26508/lsa.202302121] [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: 04/27/2023] [Revised: 08/06/2023] [Accepted: 08/07/2023] [Indexed: 08/23/2023] Open
Abstract
Loss of c-JUN leads to early mouse embryonic death, possibly because of a failure to develop a normal cardiac system. How c-JUN regulates human cardiomyocyte cell fate remains unknown. Here, we used the in vitro differentiation of human pluripotent stem cells into cardiomyocytes to study the role of c-JUN. Surprisingly, the knockout of c-JUN improved cardiomyocyte generation, as determined by the number of TNNT2+ cells. ATAC-seq data showed that the c-JUN defect led to increased chromatin accessibility on critical regulatory elements related to cardiomyocyte development. ChIP-seq data showed that the knockout c-JUN increased RBBP5 and SETD1B expression, leading to improved H3K4me3 deposition on key genes that regulate cardiogenesis. The c-JUN KO phenotype could be copied using the histone demethylase inhibitor CPI-455, which also up-regulated H3K4me3 levels and increased cardiomyocyte generation. Single-cell RNA-seq data defined three cell branches, and knockout c-JUN activated more regulons that are related to cardiogenesis. In summary, our data demonstrated that c-JUN could regulate cardiomyocyte cell fate by modulating H3K4me3 modification and chromatin accessibility and shed light on how c-JUN regulates heart development in humans.
Collapse
Affiliation(s)
- Hui Zhong
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- CAS Key Laboratory of Regenerative Biology, Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ran Zhang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Bioland Laboratory Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Guihuan Li
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ping Huang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Yudan Zhang
- Bioland Laboratory Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Jieying Zhu
- CAS Key Laboratory of Regenerative Biology, Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Junqi Kuang
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Andrew P Hutchins
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Dajiang Qin
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Bioland Laboratory Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences; Hong Kong, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease and Guangzhou Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou, China
| | - Duanqing Pei
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Dongwei Li
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
37
|
Burgon PG, Weldrick JJ, Talab OMSA, Nadeer M, Nomikos M, Megeney LA. Regulatory Mechanisms That Guide the Fetal to Postnatal Transition of Cardiomyocytes. Cells 2023; 12:2324. [PMID: 37759546 PMCID: PMC10528641 DOI: 10.3390/cells12182324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/16/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Heart disease remains a global leading cause of death and disability, necessitating a comprehensive understanding of the heart's development, repair, and dysfunction. This review surveys recent discoveries that explore the developmental transition of proliferative fetal cardiomyocytes into hypertrophic postnatal cardiomyocytes, a process yet to be well-defined. This transition is key to the heart's growth and has promising therapeutic potential, particularly for congenital or acquired heart damage, such as myocardial infarctions. Although significant progress has been made, much work is needed to unravel the complex interplay of signaling pathways that regulate cardiomyocyte proliferation and hypertrophy. This review provides a detailed perspective for future research directions aimed at the potential therapeutic harnessing of the perinatal heart transitions.
Collapse
Affiliation(s)
- Patrick G. Burgon
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar
| | - Jonathan J. Weldrick
- Department of Medicine, Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (J.J.W.); (L.A.M.)
| | | | - Muhammad Nadeer
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (O.M.S.A.T.)
| | - Michail Nomikos
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (O.M.S.A.T.)
| | - Lynn A. Megeney
- Department of Medicine, Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (J.J.W.); (L.A.M.)
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
38
|
Qu X, Hou X, Zhu K, Chen W, Chen K, Sang X, Wang C, Zhang Y, Xu H, Wang J, Hou Q, Lv L, Hou L, Zhang D. Neutrophil extracellular traps facilitate sympathetic hyperactivity by polarizing microglia toward M1 phenotype after traumatic brain injury. FASEB J 2023; 37:e23112. [PMID: 37534961 DOI: 10.1096/fj.202300752r] [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: 04/16/2023] [Revised: 06/05/2023] [Accepted: 07/13/2023] [Indexed: 08/04/2023]
Abstract
Traumatic brain injury (TBI), particularly diffuse axonal injury (DAI), often results in sympathetic hyperactivity, which can exacerbate the prognosis of TBI patients. A key component of this process is the role of neutrophils in causing neuroinflammation after TBI by forming neutrophil extracellular traps (NETs), but the connection between NETs and sympathetic excitation following TBI remains unclear. Utilizing a DAI rat model, the current investigation examined the role of NETs and the HMGB1/JNK/AP1 signaling pathway in this process. The findings revealed that sympathetic excitability intensifies and peaks 3 days post-injury, a pattern mirrored by the activation of microglia, and the escalated NETs and HMGB1 levels. Subsequent in vitro exploration validated that HMGB1 fosters microglial activation via the JNK/AP1 pathway. Moreover, in vivo experimentation revealed that the application of anti-HMGB1 and AP1 inhibitors can mitigate microglial M1 polarization post-DAI, effectively curtailing sympathetic hyperactivity. Therefore, this research elucidates that post-TBI, NETs within the PVN may precipitate sympathetic hyperactivity by stimulating M1 microglial polarization through the HMGB1/JNK/AP1 pathway.
Collapse
Affiliation(s)
- Xiaolin Qu
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoxiang Hou
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Kaixin Zhu
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
- Department of Neurosurgery, The First Naval Hospital of Southern Theater Command, Zhanjiang, China
| | - Wen Chen
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Kun Chen
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xianzheng Sang
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chenqing Wang
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yelei Zhang
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Haoxiang Xu
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Junyu Wang
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Qibo Hou
- College of Liberal Arts and Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Liquan Lv
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Lijun Hou
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Danfeng Zhang
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
39
|
Castillo-Casas JM, Caño-Carrillo S, Sánchez-Fernández C, Franco D, Lozano-Velasco E. Comparative Analysis of Heart Regeneration: Searching for the Key to Heal the Heart-Part II: Molecular Mechanisms of Cardiac Regeneration. J Cardiovasc Dev Dis 2023; 10:357. [PMID: 37754786 PMCID: PMC10531542 DOI: 10.3390/jcdd10090357] [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: 07/25/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide, among which ischemic heart disease is the most representative. Myocardial infarction results from occlusion of a coronary artery, which leads to an insufficient blood supply to the myocardium. As it is well known, the massive loss of cardiomyocytes cannot be solved due the limited regenerative ability of the adult mammalian hearts. In contrast, some lower vertebrate species can regenerate the heart after an injury; their study has disclosed some of the involved cell types, molecular mechanisms and signaling pathways during the regenerative process. In this 'two parts' review, we discuss the current state-of-the-art of the main response to achieve heart regeneration, where several processes are involved and essential for cardiac regeneration.
Collapse
Affiliation(s)
- Juan Manuel Castillo-Casas
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
| | - Sheila Caño-Carrillo
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
| | - Cristina Sánchez-Fernández
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
- Medina Foundation, 18007 Granada, Spain
| | - Diego Franco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
- Medina Foundation, 18007 Granada, Spain
| | - Estefanía Lozano-Velasco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
- Medina Foundation, 18007 Granada, Spain
| |
Collapse
|
40
|
Jia X, Lin W, Wang W. Regulation of chromatin organization during animal regeneration. CELL REGENERATION (LONDON, ENGLAND) 2023; 12:19. [PMID: 37259007 DOI: 10.1186/s13619-023-00162-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/21/2023] [Indexed: 06/02/2023]
Abstract
Activation of regeneration upon tissue damages requires the activation of many developmental genes responsible for cell proliferation, migration, differentiation, and tissue patterning. Ample evidence revealed that the regulation of chromatin organization functions as a crucial mechanism for establishing and maintaining cellular identity through precise control of gene transcription. The alteration of chromatin organization can lead to changes in chromatin accessibility and/or enhancer-promoter interactions. Like embryogenesis, each stage of tissue regeneration is accompanied by dynamic changes of chromatin organization in regeneration-responsive cells. In the past decade, many studies have been conducted to investigate the contribution of chromatin organization during regeneration in various tissues, organs, and organisms. A collection of chromatin regulators were demonstrated to play critical roles in regeneration. In this review, we will summarize the progress in the understanding of chromatin organization during regeneration in different research organisms and discuss potential common mechanisms responsible for the activation of regeneration response program.
Collapse
Affiliation(s)
- Xiaohui Jia
- National Institute of Biological Sciences, Beijing, 102206, China
- China Agricultural University, Beijing, 100083, China
| | - Weifeng Lin
- National Institute of Biological Sciences, Beijing, 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 100084, China
| | - Wei Wang
- National Institute of Biological Sciences, Beijing, 102206, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
41
|
Nguyen PD, Gooijers I, Campostrini G, Verkerk AO, Honkoop H, Bouwman M, de Bakker DEM, Koopmans T, Vink A, Lamers GEM, Shakked A, Mars J, Mulder AA, Chocron S, Bartscherer K, Tzahor E, Mummery CL, de Boer TP, Bellin M, Bakkers J. Interplay between calcium and sarcomeres directs cardiomyocyte maturation during regeneration. Science 2023; 380:758-764. [PMID: 37200435 DOI: 10.1126/science.abo6718] [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: 02/24/2022] [Accepted: 04/20/2023] [Indexed: 05/20/2023]
Abstract
Zebrafish hearts can regenerate by replacing damaged tissue with new cardiomyocytes. Although the steps leading up to the proliferation of surviving cardiomyocytes have been extensively studied, little is known about the mechanisms that control proliferation and redifferentiation to a mature state. We found that the cardiac dyad, a structure that regulates calcium handling and excitation-contraction coupling, played a key role in the redifferentiation process. A component of the cardiac dyad called leucine-rich repeat-containing 10 (Lrrc10) acted as a negative regulator of proliferation, prevented cardiomegaly, and induced redifferentiation. We found that its function was conserved in mammalian cardiomyocytes. This study highlights the importance of the underlying mechanisms required for heart regeneration and their application to the generation of fully functional cardiomyocytes.
Collapse
Affiliation(s)
- Phong D Nguyen
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Iris Gooijers
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Giulia Campostrini
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Arie O Verkerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam University Medical Center, Amsterdam, Netherlands
- Department of Experimental Cardiology, University of Amsterdam, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Hessel Honkoop
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Mara Bouwman
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Dennis E M de Bakker
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Tim Koopmans
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
- Department of Animal Physiology, Osnabrueck University, Osnabrück, Germany
| | - Aryan Vink
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Gerda E M Lamers
- Core Facility Microscopy, Institute of Biology, Leiden University, Leiden, Netherlands
| | - Avraham Shakked
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Jonas Mars
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Aat A Mulder
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Sonja Chocron
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Kerstin Bartscherer
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
- Department of Animal Physiology, Osnabrueck University, Osnabrück, Germany
| | - Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Teun P de Boer
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
- Department of Biology, University of Padua, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
- Department of Pediatric Cardiology, Division of Pediatrics, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
42
|
Ji L, Shi Y, Bian Q. Comparative genomics analyses reveal sequence determinants underlying interspecies variations in injury-responsive enhancers. BMC Genomics 2023; 24:177. [PMID: 37020217 PMCID: PMC10077677 DOI: 10.1186/s12864-023-09283-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/29/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND Injury induces profound transcriptional remodeling events, which could lead to only wound healing, partial tissue repair, or perfect regeneration in different species. Injury-responsive enhancers (IREs) are cis-regulatory elements activated in response to injury signals, and have been demonstrated to promote tissue regeneration in some organisms such as zebrafish and flies. However, the functional significances of IREs in mammals remain elusive. Moreover, whether the transcriptional responses elicited by IREs upon injury are conserved or specialized in different species, and what sequence features may underlie the functional variations of IREs have not been elucidated. RESULTS We identified a set of IREs that are activated in both regenerative and non-regenerative neonatal mouse hearts upon myocardial ischemia-induced damage by integrative epigenomic and transcriptomic analyses. Motif enrichment analysis showed that AP-1 and ETS transcription factor binding motifs are significantly enriched in both zebrafish and mouse IREs. However, the IRE-associated genes vary considerably between the two species. We further found that the IRE-related sequences in zebrafish and mice diverge greatly, with the loss of IRE inducibility accompanied by a reduction in AP-1 and ETS motif frequencies. The functional turnover of IREs between zebrafish and mice is correlated with changes in transcriptional responses of the IRE-associated genes upon injury. Using mouse cardiomyocytes as a model, we demonstrated that the reduction in AP-1 or ETS motif frequency attenuates the activation of IREs in response to hypoxia-induced damage. CONCLUSIONS By performing comparative genomics analyses on IREs, we demonstrated that inter-species variations in AP-1 and ETS motifs may play an important role in defining the functions of enhancers during injury response. Our findings provide important insights for understanding the molecular mechanisms of transcriptional remodeling in response to injury across species.
Collapse
Affiliation(s)
- Luzhang Ji
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | - Yuanyuan Shi
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | - Qian Bian
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China.
| |
Collapse
|
43
|
Zuppo DA, Missinato MA, Santana-Santos L, Li G, Benos PV, Tsang M. Foxm1 regulates cardiomyocyte proliferation in adult zebrafish after cardiac injury. Development 2023; 150:dev201163. [PMID: 36846912 PMCID: PMC10108034 DOI: 10.1242/dev.201163] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 02/13/2023] [Indexed: 03/01/2023]
Abstract
The regenerative capacity of the mammalian heart is poor, with one potential reason being that adult cardiomyocytes cannot proliferate at sufficient levels to replace lost tissue. During development and neonatal stages, cardiomyocytes can successfully divide under injury conditions; however, as these cells mature their ability to proliferate is lost. Therefore, understanding the regulatory programs that can induce post-mitotic cardiomyocytes into a proliferative state is essential to enhance cardiac regeneration. Here, we report that the forkhead transcription factor Foxm1 is required for cardiomyocyte proliferation after injury through transcriptional regulation of cell cycle genes. Transcriptomic analysis of injured zebrafish hearts revealed that foxm1 expression is increased in border zone cardiomyocytes. Decreased cardiomyocyte proliferation and expression of cell cycle genes in foxm1 mutant hearts was observed, suggesting it is required for cell cycle checkpoints. Subsequent analysis of a candidate Foxm1 target gene, cenpf, revealed that this microtubule and kinetochore binding protein is also required for cardiac regeneration. Moreover, cenpf mutants show increased cardiomyocyte binucleation. Thus, foxm1 and cenpf are required for cardiomyocytes to complete mitosis during zebrafish cardiac regeneration.
Collapse
Affiliation(s)
- Daniel A. Zuppo
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Maria A. Missinato
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
- Avidity Biosciences, 10578 Science Center Dr. Suite 125, San Diego, CA 92121, USA
| | - Lucas Santana-Santos
- Department of Computational and Systems Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Guang Li
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Panayiotis V. Benos
- Department of Computational and Systems Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Michael Tsang
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
44
|
Rolland L, Jopling C. The multifaceted nature of endogenous cardiac regeneration. Front Cardiovasc Med 2023; 10:1138485. [PMID: 36998973 PMCID: PMC10043193 DOI: 10.3389/fcvm.2023.1138485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/09/2023] [Indexed: 03/15/2023] Open
Abstract
Since the first evidence of cardiac regeneration was observed, almost 50 years ago, more studies have highlighted the endogenous regenerative abilities of several models following cardiac injury. In particular, analysis of cardiac regeneration in zebrafish and neonatal mice has uncovered numerous mechanisms involved in the regenerative process. It is now apparent that cardiac regeneration is not simply achieved by inducing cardiomyocytes to proliferate but requires a multifaceted response involving numerous different cell types, signaling pathways and mechanisms which must all work in harmony in order for regeneration to occur. In this review we will endeavor to highlight a variety of processes that have been identifed as being essential for cardiac regeneration.
Collapse
|
45
|
Gong S, McLamb F, Shea D, Vu JP, Vasquez MF, Feng Z, Bozinovic K, Hirata KK, Gersberg RM, Bozinovic G. Toxicity assessment of hexafluoropropylene oxide-dimer acid on morphology, heart physiology, and gene expression during zebrafish (Danio rerio) development. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:32320-32336. [PMID: 36462083 PMCID: PMC10017623 DOI: 10.1007/s11356-022-24542-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/25/2022] [Indexed: 05/25/2023]
Abstract
Hexafluoropropylene oxide-dimer acid (HFPO-DA) is one of the emerging replacements for the "forever" carcinogenic and toxic long-chain PFAS. HFPO-DA is a polymerization aid used for manufacturing fluoropolymers, whose global distribution and undetermined toxic properties are a concern regarding human and ecological health. To assess embryotoxic potential, zebrafish embryos were exposed to HFPO-DA at concentrations of 0.5-20,000 mg/L at 24-, 48-, and 72-h post-fertilization (hpf). Heart rate increased significantly in embryos exposed to 2 mg/L and 10 mg/L HFPO-DA across all time points. Spinal deformities and edema phenotypes were evident among embryos exposed to 1000-16,000 mg/L HFPO-DA at 72 hpf. A median lethal concentration (LC50) was derived as 7651 mg/L at 72 hpf. Shallow RNA sequencing analysis of 9465 transcripts identified 38 consistently differentially expressed genes at 0.5 mg/L, 1 mg/L, 2 mg/L, and 10 mg/L HFPO-DA exposures. Notably, seven downregulated genes were associated with visual response, and seven upregulated genes were expressed in or regulated the cardiovascular system. This study identifies biological targets and molecular pathways affected during animal development by an emerging, potentially problematic, and ubiquitous industrial chemical.
Collapse
Affiliation(s)
- Sylvia Gong
- Boz Life Science Research and Teaching Institute, San Diego, CA, USA
- Division of Extended Studies, University of California San Diego, La Jolla, CA, 92093-0355, USA
- School of Public Health, San Diego State University, San Diego, CA, USA
| | - Flannery McLamb
- Boz Life Science Research and Teaching Institute, San Diego, CA, USA
- Division of Extended Studies, University of California San Diego, La Jolla, CA, 92093-0355, USA
| | | | - Jeanne P Vu
- Boz Life Science Research and Teaching Institute, San Diego, CA, USA
- Division of Extended Studies, University of California San Diego, La Jolla, CA, 92093-0355, USA
- School of Public Health, San Diego State University, San Diego, CA, USA
| | - Miguel F Vasquez
- Boz Life Science Research and Teaching Institute, San Diego, CA, USA
- Division of Extended Studies, University of California San Diego, La Jolla, CA, 92093-0355, USA
| | - Zuying Feng
- Boz Life Science Research and Teaching Institute, San Diego, CA, USA
- School of Public Health, San Diego State University, San Diego, CA, USA
| | - Kesten Bozinovic
- Boz Life Science Research and Teaching Institute, San Diego, CA, USA
- Division of Extended Studies, University of California San Diego, La Jolla, CA, 92093-0355, USA
- Graduate School of Arts and Sciences, Georgetown University, Washington, DC, USA
| | - Ken K Hirata
- Boz Life Science Research and Teaching Institute, San Diego, CA, USA
- Division of Extended Studies, University of California San Diego, La Jolla, CA, 92093-0355, USA
| | | | - Goran Bozinovic
- Boz Life Science Research and Teaching Institute, San Diego, CA, USA.
- School of Public Health, San Diego State University, San Diego, CA, USA.
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, 92093-0355, USA.
| |
Collapse
|
46
|
Singh BN, Yucel D, Garay BI, Tolkacheva EG, Kyba M, Perlingeiro RCR, van Berlo JH, Ogle BM. Proliferation and Maturation: Janus and the Art of Cardiac Tissue Engineering. Circ Res 2023; 132:519-540. [PMID: 36795845 PMCID: PMC9943541 DOI: 10.1161/circresaha.122.321770] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
During cardiac development and morphogenesis, cardiac progenitor cells differentiate into cardiomyocytes that expand in number and size to generate the fully formed heart. Much is known about the factors that regulate initial differentiation of cardiomyocytes, and there is ongoing research to identify how these fetal and immature cardiomyocytes develop into fully functioning, mature cells. Accumulating evidence indicates that maturation limits proliferation and conversely proliferation occurs rarely in cardiomyocytes of the adult myocardium. We term this oppositional interplay the proliferation-maturation dichotomy. Here we review the factors that are involved in this interplay and discuss how a better understanding of the proliferation-maturation dichotomy could advance the utility of human induced pluripotent stem cell-derived cardiomyocytes for modeling in 3-dimensional engineered cardiac tissues to obtain truly adult-level function.
Collapse
Affiliation(s)
- Bhairab N. Singh
- Department of Pediatrics, University of Minnesota, MN, USA
- Department of Biomedical Engineering, University of Minnesota, MN, USA
- Stem Cell Institute, University of Minnesota, MN, USA
| | - Dogacan Yucel
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Bayardo I. Garay
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
- Medical Scientist Training Program, University of Minnesota Medical School, MN, USA
| | - Elena G. Tolkacheva
- Department of Biomedical Engineering, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
- Institute for Engineering in Medicine, University of Minnesota, MN, USA
| | - Michael Kyba
- Department of Pediatrics, University of Minnesota, MN, USA
- Stem Cell Institute, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Rita C. R. Perlingeiro
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Jop H. van Berlo
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Brenda M. Ogle
- Department of Pediatrics, University of Minnesota, MN, USA
- Department of Biomedical Engineering, University of Minnesota, MN, USA
- Stem Cell Institute, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
- Institute for Engineering in Medicine, University of Minnesota, MN, USA
- Masonic Cancer Center, University of Minnesota, MN, USA
| |
Collapse
|
47
|
Kraus L, Beavens B. The Current Therapeutic Role of Chromatin Remodeling for the Prognosis and Treatment of Heart Failure. Biomedicines 2023; 11:biomedicines11020579. [PMID: 36831115 PMCID: PMC9953583 DOI: 10.3390/biomedicines11020579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Cardiovascular diseases are a major cause of death globally, with no cure to date. Many interventions have been studied and suggested, of which epigenetics and chromatin remodeling have been the most promising. Over the last decade, major advancements have been made in the field of chromatin remodeling, particularly for the treatment of heart failure, because of innovations in bioinformatics and gene therapy. Specifically, understanding changes to the chromatin architecture have been shown to alter cardiac disease progression via variations in genomic sequencing, targeting cardiac genes, using RNA molecules, and utilizing chromatin remodeler complexes. By understanding these chromatin remodeling mechanisms in an injured heart, treatments for heart failure have been suggested through individualized pharmaceutical interventions as well as biomarkers for major disease states. By understanding the current roles of chromatin remodeling in heart failure, a potential therapeutic approach may be discovered in the future.
Collapse
|
48
|
RING Finger Protein 10 Regulates AP-1/Meox2 to Mediate Pirarubicin-Induced Cardiomyocyte Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:7872193. [PMID: 36713029 PMCID: PMC9883094 DOI: 10.1155/2023/7872193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/12/2022] [Accepted: 11/25/2022] [Indexed: 01/21/2023]
Abstract
Pirarubicin (THP) is one of the classic chemotherapy drugs for cancer treatment. It is often clinically limited because of its cardiotoxicity. The occurrence and development of THP-mediated chemotherapy-related cardiotoxicity (CRC) may be reversed by RING finger protein 10 (RNF10). This study was performed with the aim of evaluating the inhibitory effect of RNF10 on THP-mediated CRC and its molecular mechanism. In vivo, we found that the expression of RNF10 decreased in THP-induced CRC rats, accompanied by Meox2 inhibition and AP-1 activation, resulting in increased cardiomyocyte apoptosis. After small interfering RNA (siRNA) and lentivirus transfection (Lv) of RNF10 in vitro, the expression of RNF10, Meox2, and AP-1 proteins and the degree of cardiomyocyte apoptosis were detected. We found that overexpression of RNF10 in H9C2 cardiomyocytes significantly promoted Meox2 and inhibited AP-1, alleviated apoptosis, and showed further inhibitory activity on THP-induced cardiomyocyte toxicity. Silencing RNF10 showed the opposite result. Our study showed that RNF10 inhibited THP-induced CRC through the activity of Meox2 and AP-1 proteins. RNF10 may be the next drug target for the treatment of CRC and other related cardiovascular diseases.
Collapse
|
49
|
Zhang J, Ouyang Z, Xia L, Wang Q, Zheng F, Xu K, Xing Y, Wei K, Shi S, Li C, Yang J. Dynamic chromatin landscape encodes programs for perinatal transition of cardiomyocytes. Cell Death Dis 2023; 9:11. [PMID: 36653336 PMCID: PMC9849264 DOI: 10.1038/s41420-023-01322-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/19/2023]
Abstract
The perinatal period occurring immediately before and after birth is critical for cardiomyocytes because they must change rapidly to accommodate the switch from fetal to neonatal circulation after birth. This transition is a well-orchestrated process, and any perturbation leads to unhealthy cardiomyocytes and heart disease. Despite its importance, little is known about how this transition is regulated and controlled. Here, by mapping the genome-wide chromatin accessibility, transcription-centered long-range chromatin interactions and gene expression in cardiomyocytes undergoing perinatal transition, we discovered two key transcription factors, MEF2 and AP1, that are crucial for driving the phenotypic changes within the perinatal window. Thousands of dynamic regulatory elements were found in perinatal cardiomyocytes and we show these elements mediated the transcriptional reprogramming through an elegant chromatin high-order architecture. We recompiled transcriptional program of induced stem cell-derived cardiomyocytes according to our discovered network, and they showed adult cardiomyocyte-like electrophysiological expression. Our work provides a comprehensive regulatory resource of cardiomyocytes perinatal reprogramming, and aids the gap-filling of cardiac translational research.
Collapse
Affiliation(s)
- Jing Zhang
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China ,grid.41156.370000 0001 2314 964XJiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China
| | - Zhaohui Ouyang
- grid.24516.340000000123704535Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 200092 Shanghai, China
| | - Limei Xia
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China ,grid.41156.370000 0001 2314 964XJiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China
| | - Qi Wang
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China ,grid.41156.370000 0001 2314 964XJiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China
| | - Feng Zheng
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China ,grid.41156.370000 0001 2314 964XJiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China
| | - Kun Xu
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China ,grid.41156.370000 0001 2314 964XJiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China
| | - Yuexian Xing
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China
| | - Ke Wei
- grid.24516.340000000123704535Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 200092 Shanghai, China
| | - Shaolin Shi
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China
| | - Chaojun Li
- grid.89957.3a0000 0000 9255 8984State Key Laboratory of Reproductive Medicine and China International Joint Research Center on Environment and Human Health, Center for Global Health, School of Public Health, Gusu School, Nanjing Medical University, 211166 Nanjing, China
| | - Jingping Yang
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China ,grid.41156.370000 0001 2314 964XJiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093 Nanjing, Jiangsu China
| |
Collapse
|
50
|
Tanaka EM. Now that We Got There, What Next? Methods Mol Biol 2023; 2562:471-479. [PMID: 36272095 DOI: 10.1007/978-1-0716-2659-7_31] [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] [Indexed: 06/16/2023]
Abstract
As seen in the protocols in this book, the opportunities to pursue work at the cellular and molecular work in salamanders have considerably broadened over the last years. The availability of genomic information and genome editing, and the possibility to image tissues live and other methods enhance the spectrum of biological questions accessible to all researchers. Here I provide a personal perspective on what I consider exciting future questions open for investigation.
Collapse
Affiliation(s)
- Elly M Tanaka
- Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria.
| |
Collapse
|