1
|
Li L, Coarfa C, Yuan Y, Abu-Taha I, Wang X, Song J, Zeng Y, Chen X, Koirala A, Grimm SL, Kamler M, McClendon LK, Tallquist M, Nattel S, Dobrev D, Li N. Fibroblast-Restricted Inflammasome Activation Promotes Atrial Fibrillation and Heart Failure With Diastolic Dysfunction. JACC Basic Transl Sci 2025:S2452-302X(25)00061-0. [PMID: 40243956 DOI: 10.1016/j.jacbts.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 01/31/2025] [Accepted: 02/01/2025] [Indexed: 04/18/2025]
Abstract
Atrial fibrillation (AF) often coexists with heart failure, both involving inflammatory signaling and cardiac fibroblasts. To understand the role of fibroblast NLR family pyrin domain containing 3 (NLRP3) inflammasome in cardiac function, we found that NLRP3 was up-regulated in atrial fibroblasts from AF patients. Fibroblast-specific activation of NLRP3 in mice induced AF-promoting atrial myopathy and heart failure with diastolic dysfunction, accompanied by increased fibrosis, and reduced conduction velocity. Knockdown of NLRP3 prevented the AF-promoting atrial substrate and cardiomyopathy in the context of NLRP3 activation in fibroblasts. We identify the fibroblast NLRP3 inflammasome as a key pathway governing the promotion of proarrhythmic fibrosis in AF and cardiomyopathy.
Collapse
Affiliation(s)
- Luge Li
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, Texas, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Yue Yuan
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, Texas, USA
| | - Issam Abu-Taha
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany
| | - Xiaolei Wang
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, Texas, USA
| | - Jia Song
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, Texas, USA
| | - Yuying Zeng
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, Texas, USA
| | - Xiaohui Chen
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, Texas, USA
| | - Amrit Koirala
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Sandra L Grimm
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Markus Kamler
- Department of Thoracic and Cardiovascular Surgery, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Lisa K McClendon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Michelle Tallquist
- Center for Cardiovascular Research, University of Hawaii, Honolulu, Hawaii, USA
| | - Stanley Nattel
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany; Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada; IHU LIRYC and Fondation Bordeaux Université, Bordeaux, France
| | - Dobromir Dobrev
- Institute of Pharmacology, University Duisburg-Essen, Essen, Germany; Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada; Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
| | - Na Li
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, Texas, USA; Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas, USA.
| |
Collapse
|
2
|
Li L, Coarfa C, Yuan Y, Abu-Taha I, Wang X, Song J, Koirala A, Grimm SL, Kamler M, Mullany LK, Tallquist M, Nattel S, Dobrev D, Li N. Fibroblast-specific inflammasome activation predisposes to atrial fibrillation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.18.541326. [PMID: 37292708 PMCID: PMC10245773 DOI: 10.1101/2023.05.18.541326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Background Recent work has shown that the NLR-family-pyrin-domain-containing 3 (NLRP3) inflammasome is expressed in cardiomyocytes and when specifically activated causes atrial electrical remodeling and arrhythmogenicity. Whether the NLRP3-inflammasome system is functionally important in cardiac fibroblasts (FBs) remains controversial. In this study, we sought to uncover the potential contribution of FB NLRP3-inflammasome signaling to the control of cardiac function and arrhythmogenesis. Methods Digital-PCR was performed to determine the expression of NLRP3-pathway components in FBs isolated from human biopsy samples of AF and sinus rhythm patients. NLRP3-system protein expression was determined by immunoblotting in atria of canines with electrically maintained AF. Using the inducible, resident fibroblast (FB)-specific Tcf21-promoter-Cre system (Tcf21iCre as control), we established a FB-specific knockin (FB-KI) mouse model with FB-restricted expression of constitutively active NLRP3. Cardiac function and arrhythmia susceptibility in mice were assessed by echocardiography, programmed electrical stimulation, and optical mapping studies. Results NLRP3 and IL1B were upregulated in atrial FBs of patients with persistent AF. Protein levels of NLRP3, ASC, and pro-Interleukin-1β were increased in atrial FBs of a canine AF model. Compared with the control mice, FB-KI mice exhibited enlarged left atria (LA) and reduced LA contractility, a common determinant of AF. The FBs from FB-KI mice were more transdifferentiated, migratory, and proliferative compared to the FBs from control mice. FB-KI mice showed increased cardiac fibrosis, atrial gap junction remodeling, and reduced conduction velocity, along with increased AF susceptibility. These phenotypic changes were supported by single nuclei (sn)RNA-seq analysis, which revealed enhanced extracellular matrix remodeling, impaired communication among cardiomyocytes, and altered metabolic pathways across multiple cell types. Conclusions Our results show that the FB-restricted activation of the NLRP3-inflammasome system leads to fibrosis, atrial cardiomyopathy, and AF. Activation of NLRP3-inflammasome in resident FBs exhibits cell-autonomous function by increasing the activity of cardiac FBs, fibrosis, and connexin remodeling. This study establishes the NLRP3-inflammasome as a novel FB-signaling pathway contributing to AF pathogenesis.
Collapse
|
3
|
Song BW, Kim S, Kim R, Jeong S, Moon H, Kim H, Vasileva EA, Mishchenko NP, Fedoreyev SA, Stonik VA, Lee MY, Kim J, Kim HK, Han J, Chang W. Regulation of Inflammation-Mediated Endothelial to Mesenchymal Transition with Echinochrome a for Improving Myocardial Dysfunction. Mar Drugs 2022; 20:756. [PMID: 36547903 PMCID: PMC9781361 DOI: 10.3390/md20120756] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Endothelial-mesenchymal transition (EndMT) is a process by which endothelial cells (ECs) transition into mesenchymal cells (e.g., myofibroblasts and smooth muscle cells) and induce fibrosis of cells/tissues, due to ischemic conditions in the heart. Previously, we reported that echinochrome A (EchA) derived from sea urchin shells can modulate cardiovascular disease by promoting anti-inflammatory and antioxidant activity; however, the mechanism underlying these effects was unclear. We investigated the role of EchA in the EndMT process by treating human umbilical vein ECs (HUVECs) with TGF-β2 and IL-1β, and confirmed the regulation of cell migration, inflammatory, oxidative responses and mitochondrial dysfunction. Moreover, we developed an EndMT-induced myocardial infarction (MI) model to investigate the effect of EchA in vivo. After EchA was administered once a day for a total of 3 days, the histological and functional improvement of the myocardium was investigated to confirm the control of the EndMT. We concluded that EchA negatively regulates early or inflammation-related EndMT and reduces the myofibroblast proportion and fibrosis area, meaning that it may be a potential therapy for cardiac regeneration or cardioprotection from scar formation and cardiac fibrosis due to tissue granulation. Our findings encourage the study of marine bioactive compounds for the discovery of new therapeutics for recovering ischemic cardiac injuries.
Collapse
Affiliation(s)
- Byeong-Wook Song
- Institute for Bio-Medical Convergence, Catholic Kwandong University International St. Mary’s Hospital, Incheon 22711, Republic of Korea
| | - Sejin Kim
- Department of Biology Education, College of Education, Pusan National University, Busan 46241, Republic of Korea
| | - Ran Kim
- Department of Biology Education, College of Education, Pusan National University, Busan 46241, Republic of Korea
| | - Seongtae Jeong
- Institute for Bio-Medical Convergence, Catholic Kwandong University International St. Mary’s Hospital, Incheon 22711, Republic of Korea
| | - Hanbyeol Moon
- Institute for Bio-Medical Convergence, Catholic Kwandong University International St. Mary’s Hospital, Incheon 22711, Republic of Korea
| | - Hojin Kim
- Institute for Bio-Medical Convergence, Catholic Kwandong University International St. Mary’s Hospital, Incheon 22711, Republic of Korea
| | - Elena A. Vasileva
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia
| | - Natalia P. Mishchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia
| | - Sergey A. Fedoreyev
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia
| | - Valentin A. Stonik
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, 690022 Vladivostok, Russia
| | - Min Young Lee
- Department of Molecular Physiology, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jongmin Kim
- Department of Life Systems, Sookmyung Women’s University, Seoul 04310, Republic of Korea
| | - Hyoung Kyu Kim
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutic Center, Inje University, Busan 47392, Republic of Korea
| | - Jin Han
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutic Center, Inje University, Busan 47392, Republic of Korea
| | - Woochul Chang
- Department of Biology Education, College of Education, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
4
|
Mazurara GR, Dallagnol JCC, Chatenet D, Allen BG, Hébert TE. The complicated lives of GPCRs in cardiac fibroblasts. Am J Physiol Cell Physiol 2022; 323:C813-C822. [PMID: 35938678 DOI: 10.1152/ajpcell.00120.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The role of different G protein-coupled receptors (GPCRs) in the cardiovascular system is well understood in cardiomyocytes and vascular smooth muscle cells (VSMCs). In the former, stimulation of Gs-coupled receptors leads to increases in contractility, while stimulation of Gq-coupled receptors modulates cellular survival and hypertrophic responses. In VSMCs, stimulation of GPCRs also modulates contractile and cell growth phenotypes. Here, we will focus on the relatively less well studied effects of GPCRs in cardiac fibroblasts, focusing on key signalling events involved in the activation and differentiation of these cells. We also review the hierarchy of signalling events driving the fibrotic response and the communications between fibroblasts and other cells in the heart. We discuss how such events may be distinct depending on where the GPCRs and their associated signalling machinery are localized in these cells with an emphasis on nuclear membrane-localized receptors. Finally, we explore what such connections between cell surface and nuclear GPCR signalling might mean for cardiac fibrosis.
Collapse
Affiliation(s)
- Grace R Mazurara
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | - Juliana C C Dallagnol
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada.,Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Laval, Québec, Canada.,Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
| | - David Chatenet
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Laval, Québec, Canada
| | - Bruce G Allen
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| |
Collapse
|
5
|
Perreault LR, Le TT, Oudin MJ, Black LD. RNA sequencing indicates age-dependent shifts in the cardiac fibroblast transcriptome between fetal, neonatal, and adult developmental ages. Physiol Genomics 2021; 53:414-429. [PMID: 34281425 PMCID: PMC8560366 DOI: 10.1152/physiolgenomics.00074.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/16/2021] [Indexed: 11/22/2022] Open
Abstract
Cardiac fibroblasts are responsible for extracellular matrix turnover and repair in the cardiac environment and serve to help facilitate immune responses. However, it is well established that they have a significant phenotypic heterogeneity with respect to location, physiological conditions, and developmental age. The goal of this study was to provide an in-depth transcriptomic profile of cardiac fibroblasts derived from rat hearts at fetal, neonatal, and adult developmental ages to ascertain variations in gene expression that may drive functional differences in these cells at these specific stages of development. We performed RNA sequencing (RNA-seq) of cardiac fibroblasts isolated from fetal, neonatal, and adult rats and compared with the rat genome. Principal component analysis of RNA-seq data suggested that data variance was predominantly due to developmental age. Differential expression and gene set enrichment analysis against Gene Ontology and Kyoto Encyclopedia of Genes and Genomes datasets indicated an array of differences across developmental ages, including significant decreases in cardiac development and cardiac function-associated genes with age and a significant increase in immune- and inflammatory-associated functions, particularly immune cell signaling and cytokine and chemokine production, with respect to increasing developmental age. These results reinforce established evidence of diverse phenotypic heterogeneity of fibroblasts with respect to developmental age. Furthermore, based on our analysis of gene expression, age-specific alterations in cardiac fibroblasts may play a crucial role in observed differences in cardiac inflammation and immune response observed across developmental ages.
Collapse
Affiliation(s)
- Luke R Perreault
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Thanh T Le
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Madeleine J Oudin
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
- Cellular, Molecular, and Developmental Biology Program, Tufts Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts
| | - Lauren D Black
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
- Cellular, Molecular, and Developmental Biology Program, Tufts Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts
| |
Collapse
|
6
|
Zhang SQ, Bao YN, Lv LY, Du XH, Wang YC. Conophylline Suppresses Angiotensin II-Induced Myocardial Fibrosis In Vitro via the BMP4/JNK Pathway. Bull Exp Biol Med 2021; 171:305-311. [PMID: 34302205 DOI: 10.1007/s10517-021-05217-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Indexed: 11/29/2022]
Abstract
We studied the effects and mechanisms of action of conophylline in different concentrations in the original in vitro model of myocardial fibrosis (treatment of cardiac fibroblasts isolated form the hearts of newborn rats with angiotensin II). Viability, collagen content, and expression of related protein in cardiac fibroblasts were assessed using the MTT-test, Sircol assay, and Western blotting, respectively. Conophylline markedly protected the cultured cells against the development of angiotensin II-induced fibrosis, which was seen from reduced viability of fibroblasts, decreased collagen content, and down-regulation of the expression of α-smooth muscle actin (α-SMA). Conophylline did not affect the TGF-β pathway altered by angiotensin II, but markedly decreased the level of bone morphogenetic protein-4 (BMP4) enhanced by angiotensin II and BMP4 itself. Conophylline produced no effect on phosphorylation of α-SMA and Smad homologue-1/5/8, the classic BMP4 downstream pathway elements, but reduced the level of c-Jun N-terminal kinase (JNK) elevated by BMP4. Conophylline did not inhibit the development of myocardial fibrosis in the presence of JNK activator anisomycin. Thus, conophylline inhibited angiotensin II-provoked myocardial fibrosis via the BMP4/JNK pathway.
Collapse
Affiliation(s)
- S Q Zhang
- Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Y N Bao
- Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - L Y Lv
- Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - X H Du
- Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Y C Wang
- Qiqihar Medical University, Qiqihar, Heilongjiang, China.
| |
Collapse
|
7
|
Liu M, López de Juan Abad B, Cheng K. Cardiac fibrosis: Myofibroblast-mediated pathological regulation and drug delivery strategies. Adv Drug Deliv Rev 2021; 173:504-519. [PMID: 33831476 PMCID: PMC8299409 DOI: 10.1016/j.addr.2021.03.021] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 02/16/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023]
Abstract
Cardiac fibrosis remains an unresolved problem in heart diseases. After initial injury, cardiac fibroblasts (CFs) are activated and subsequently differentiate into myofibroblasts (myoFbs) that are major mediator cells in the pathological remodeling. MyoFbs exhibit proliferative and secretive characteristics, and contribute to extracellular matrix (ECM) turnover, collagen deposition. The persistent functions of myoFbs lead to fibrotic scars and cardiac dysfunction. The anti-fibrotic treatment is hindered by the elusive mechanism of fibrosis and lack of specific targets on myoFbs. In this review, we will outline the progress of cardiac fibrosis and its contributions to the heart failure. We will also shed light on the role of myoFbs in the regulation of adverse remodeling. The communication between myoFbs and other cells that are involved in the heart injury and repair respectively will be reviewed in detail. Then, recently developed therapeutic strategies to treat fibrosis will be summarized such as i) chimeric antigen receptor T cell (CAR-T) therapy with an optimal target on myoFbs, ii) direct reprogramming from stem cells to quiescent CFs, iii) "off-target" small molecular drugs. The application of nano/micro technology will be discussed as well, which is involved in the construction of cell-based biomimic platforms and "pleiotropic" drug delivery systems.
Collapse
Affiliation(s)
- Mengrui Liu
- Department of Molecular Biomedical Sciences, North Carolina State University, NC, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA
| | - Blanca López de Juan Abad
- Department of Molecular Biomedical Sciences, North Carolina State University, NC, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences, North Carolina State University, NC, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA.
| |
Collapse
|
8
|
Reichardt IM, Robeson KZ, Regnier M, Davis J. Controlling cardiac fibrosis through fibroblast state space modulation. Cell Signal 2020; 79:109888. [PMID: 33340659 DOI: 10.1016/j.cellsig.2020.109888] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/13/2022]
Abstract
The transdifferentiation of cardiac fibroblasts into myofibroblasts after cardiac injury has traditionally been defined by a unidirectional continuum from quiescent fibroblasts, through activated fibroblasts, and finally to fibrotic-matrix producing myofibroblasts. However, recent lineage tracing and single cell RNA sequencing experiments have demonstrated that fibroblast transdifferentiation is much more complex. Growing evidence suggests that fibroblasts are more heterogenous than previously thought, and many new cell states have recently been identified. This review reexamines conventional fibroblast transdifferentiation paradigms with a dynamic state space lens, which could enable a more complex understanding of how fibroblast state dynamics alters fibrotic remodeling of the heart. This review will define cellular state space, how it relates to fibroblast state transitions, and how the canonical and non-canonical fibrotic signaling pathways modulate fibroblast cell state and cardiac fibrosis. Finally, this review explores the therapeutic potential of fibroblast state space modulation by p38 inhibition, yes-associated protein (YAP) inhibition, and fibroblast reprogramming.
Collapse
Affiliation(s)
- Isabella M Reichardt
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States.
| | - Kalen Z Robeson
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States.
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States; Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA 98109, United States; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, United States; Center for Translational Muscle Research, University of Washington, Seattle, WA 98109, United States.
| | - Jennifer Davis
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States; Department of Pathology, University of Washington, 850 Republican, #343, Seattle, WA 98109, United States; Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA 98109, United States; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, United States; Center for Translational Muscle Research, University of Washington, Seattle, WA 98109, United States.
| |
Collapse
|
9
|
Vazir A, Fox K, Westaby J, Evans MJ, Westaby S. Can we remove scar and fibrosis from adult human myocardium? Eur Heart J 2020; 40:960-966. [PMID: 30203057 DOI: 10.1093/eurheartj/ehy503] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/09/2018] [Accepted: 09/04/2018] [Indexed: 12/12/2022] Open
Abstract
The pathological processes leading to heart failure are characterized by the formation of fibrosis and scar, yet the dynamics of scar production and removal are incompletely understood. Spontaneous disappearance of myocardial collagen is reported in infancy but doubted in adulthood where scar volume constitutes a better prognostic indicator than the conventional parameters of ventricular function. Whilst certain drugs are known to attenuate myocardial fibrosis evidence is emerging that stem cell therapy also has the potential to reduce scar size and improve myocardial viability. Both animal studies and clinical trials support the concept that, as in infancy, cellular processes can be triggered to remove collagen and regenerate injured myocardium. The molecular mechanisms likely involve anti-fibrotic cytokines growth factors and matrix-metalloproteinases. Autologous cardiac, bone-marrow and adipose tissue derived stem cells have each shown efficacy. Specific immune privileged mesenchymal stem cells and genetically modified immunomodulatory progenitor cells may in turn provide an allogenic source for the paracrine effects. Thus autologous and allogenic cells both have the potential through paracrine action to reduce scar volume, boost angiogenesis and improve ventricular morphology. The potential benefit of myocardial cell therapy for routine treatment of heart failure is an area that requires further study.
Collapse
Affiliation(s)
- Ali Vazir
- National Heart and Lung Institute, Imperial College London and ICMS, Royal Brompton Hospital, Dovehouse Street, London, UK
| | - Kim Fox
- National Heart and Lung Institute, Imperial College London and ICMS, Royal Brompton Hospital, Dovehouse Street, London, UK
| | - Joseph Westaby
- Department of Pathology, St George's University Hospital NHS Foundation Trust, Blackshaw Road, Tooting, London, UK
| | - Martin J Evans
- School of Biosciences, Cardiff University, The Sir Martin Evans Building, Museum Avenue, Cardiff, Wales, UK
| | - Stephen Westaby
- National Heart and Lung Institute, Imperial College London and ICMS, Royal Brompton Hospital, Dovehouse Street, London, UK.,Institute of Life Science, Swansea University, Singleton Park, Swansea, Wales, UK
| |
Collapse
|
10
|
Lu H, Chen R, Barnie PA, Tian Y, Zhang S, Xu H, Chakrabarti S, Su Z. Fibroblast transdifferentiation promotes conversion of M1 macrophages and replenishment of cardiac resident macrophages following cardiac injury in mice. Eur J Immunol 2020; 50:795-808. [PMID: 32068249 DOI: 10.1002/eji.201948414] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 02/13/2020] [Accepted: 02/17/2020] [Indexed: 12/24/2022]
Abstract
Resident cardiac macrophages play important roles in homeostasis, maintenance of cardiac function, and tissue repair. After cardiac injury, monocytes infiltrate the tissue, undergo phenotypic and functional changes, and are involved in inflammatory injury and functional remodelling. However, the fate of cardiac infiltrating/polarized macrophages and the relationship between these cells and resident cardiac macrophage replenishment following injury remain unclear. Our results showed that angiotensin II induces cardiac fibroblast transdifferentiation into cardiac myofibroblasts (MFBs). In cocultures with MFBs and murine macrophages, the MFBs promoted macrophage polarization to M1 phenotype, followed by selective apoptosis, which was associated with TNF/TNFR1 axis and independent of NO production. Surprisingly, after 36 h of coculture, the surviving macrophages were converted to M2 phenotype and settled in heart, which was dependent on leptin produced by MFBs or polarized macrophages via the PI3K or Akt pathway. CCR2+ CD45.2+ cells adoptively transferred into CD45.1+ mice with viral myocarditis, differentiated into CD45.2+ CCR2+ CX3CR1+ M2 cells during the resolution of inflammation and settled within the heart. Our data highlight a novel mechanism related to the renewal or replenishment of cardiac resident macrophages following cardiac injury; and suggest that transdifferentiation of cardiac fibroblasts may promote the resolution of inflammation.
Collapse
Affiliation(s)
- Hongxiang Lu
- International Genome Center, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu University, Zhenjiang, China
| | - Rong Chen
- International Genome Center, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu University, Zhenjiang, China
| | | | - Yu Tian
- International Genome Center, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu University, Zhenjiang, China
| | - Shiqing Zhang
- International Genome Center, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu University, Zhenjiang, China
| | - Huaxi Xu
- Department of Immunology, Jiangsu University, Zhenjiang, China
| | - Subrata Chakrabarti
- Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu University, Zhenjiang, China.,Laboratory Center, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
11
|
Acetaldehyde dehydrogenase 2 deficiency exacerbates cardiac fibrosis by promoting mobilization and homing of bone marrow fibroblast progenitor cells. J Mol Cell Cardiol 2019; 137:107-118. [PMID: 31668970 DOI: 10.1016/j.yjmcc.2019.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/22/2022]
Abstract
Cardiac fibrosis is a common feature of various cardiovascular diseases. Previous studies showed that acetaldehyde dehydrogenase 2 (ALDH2) deficiency exacerbated pressure overload-induced heart failure. However, the role and mechanisms of cardiac fibrosis in this process remain largely unknown. This study aimed to investigate the effect of ALDH2 deficiency on cardiac fibrosis in transverse aortic constriction (TAC) induced pressure overload model in mice. Echocardiography and histological analysis revealed cardiac dysfunction and enhanced cardiac fibrosis in TAC-operated animals; ALDH2 deficiency further aggravated these changes. ALDH2 chimeric mice were generated by bone marrow (BM) transplantation of WT mice into the lethally irradiated ALDH2KO mice. The proportion of circulating fibroblast progenitor cells (FPCs) and ROS level in BM after TAC were significantly higher in ALDH2KO mice than in ALDH2 chimeric mice. Furthermore, FPCs were isolated and cultured for in vitro mechanistic studies. The results showed that the stem cell-derived factor 1 (SDF-1)/C-X-C chemokine receptor 4 (CXCR4) axis played a major role in the recruitment of FPCs. In conclusion, our research reveals that increased bone marrow FPCs mobilization and myocardial homing contribute to the enhanced cardiac fibrosis and dysfunction induced by TAC in ALDH2 KO mice via exacerbating accumulation of ROS in BM and myocardial SDF-1 expression.
Collapse
|
12
|
Gao L, Yang L, Wang L, Geng Z, Wei Y, Gourley G, Zhang J. Relationship Between the Efficacy of Cardiac Cell Therapy and the Inhibition of Differentiation of Human iPSC-Derived Nonmyocyte Cardiac Cells Into Myofibroblast-Like Cells. Circ Res 2019; 123:1313-1325. [PMID: 30566050 DOI: 10.1161/circresaha.118.313094] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
RATIONALE Myofibroblasts are believed to evolve from precursor cells; however, whether noncardiomyocyte cardiac cells (NMCCs; ie, endothelial cells, smooth muscle cells, pericytes, and fibroblasts) that have been derived from human-induced pluripotent stem cells (hiPSCs) can transdifferentiate into myofibroblast-like cells, and if so, whether this process reduces the efficacy of hiPSC-NMCC therapy, is unknown. OBJECTIVE To determine whether hiPSC-NMCCs can differentiate to myofibroblast-like cells and whether limiting the transdifferentiation of hiPSC-NMCCs can improve their effectiveness for myocardial repair. METHODS AND RESULTS When endothelial cells, smooth muscle cells, pericytes, and fibroblasts that had been generated from hiPSCs were cultured with TGF-β (transforming growth factor-β), the expression of myofibroblast markers increased, whereas endothelial cell, smooth muscle cell, pericyte, and fibroblast marker expression declined. TGF-β-associated myofibroblast differentiation was accompanied by increases in the signaling activity of Smad, Snail, and mTOR (mammalian target of rapamycin). However, measures of pathway activation, proliferation, apoptosis, migration, and protein expression in hiPSC-endothelial cell-derived, smooth muscle cell-derived, pericyte-derived, and fibroblast-derived myofibroblast-like cells differed. Furthermore, when hiPSC-NMCCs were transplanted into the hearts of mice after myocardial infarction, ≈21% to 35% of the transplanted hiPSC-NMCCs expressed myofibroblast markers 1 week later, compared with <7% of transplanted cells ( P<0.01, each cell type) in animals that were treated with both hiPSC-NMCCs and the TGF-β inhibitor galunisertib. Galunisertib coadministration was also associated with significant improvements in fibrotic area, left ventricular dilatation, vascular density, and cardiac function. CONCLUSIONS hiPSC-NMCCs differentiate into myofibroblast-like cells when cultured with TGF-β or when transplanted into infarcted mouse hearts, and the phenotypes of the myofibroblast-like cells can differ depending on the lineage of origin. TGF-β inhibition significantly improved the efficacy of transplanted hiPSC-NMCCs for cardiac repair, perhaps by limiting the differentiation of hiPSC-NMCCs into myofibroblast-like cells.
Collapse
Affiliation(s)
- Ling Gao
- From the Department of Biomedical Engineering, University of Alabama at Birmingham (L.G., L.W., Y.W., J.Z.)
| | - Libang Yang
- Department of Medicine (L.Y., Z.G.), University of Minnesota, Minneapolis
| | - Lu Wang
- From the Department of Biomedical Engineering, University of Alabama at Birmingham (L.G., L.W., Y.W., J.Z.)
| | - Zhaohui Geng
- Department of Medicine (L.Y., Z.G.), University of Minnesota, Minneapolis
| | - Yuhua Wei
- From the Department of Biomedical Engineering, University of Alabama at Birmingham (L.G., L.W., Y.W., J.Z.)
| | - Glenn Gourley
- Department of Pediatrics (G.G.), University of Minnesota, Minneapolis (G.G.)
| | - Jianyi Zhang
- From the Department of Biomedical Engineering, University of Alabama at Birmingham (L.G., L.W., Y.W., J.Z.)
| |
Collapse
|
13
|
Klopsch C, Skorska A, Ludwig M, Lemcke H, Maass G, Gaebel R, Beyer M, Lux C, Toelk A, Müller K, Maschmeier C, Rohde S, Mela P, Müller-Hilke B, Jockenhoevel S, Vollmar B, Jaster R, David R, Steinhoff G. Intramyocardial angiogenetic stem cells and epicardial erythropoietin save the acute ischemic heart. Dis Model Mech 2018; 11:dmm.033282. [PMID: 29752300 PMCID: PMC6031356 DOI: 10.1242/dmm.033282] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 04/26/2018] [Indexed: 12/14/2022] Open
Abstract
Ischemic heart failure is the leading cause of mortality worldwide. An early boost of intracardiac regenerative key mechanisms and angiogenetic niche signaling in cardiac mesenchymal stem cells (MSCs) could improve myocardial infarction (MI) healing. Epicardial erythropoietin (EPO; 300 U kg-1) was compared with intraperitoneal and intramyocardial EPO treatments after acute MI in rats (n=156). Real-time PCR and confocal microscopy revealed that epicardial EPO treatment enhanced levels of intracardiac regenerative key indicators (SDF-1, CXCR4, CD34, Bcl-2, cyclin D1, Cdc2 and MMP2), induced transforming growth factor β (TGF-β)/WNT signaling in intramyocardial MSC niches through the direct activation of AKT and upregulation of upstream signals FOS and Fzd7, and augmented intracardiac mesenchymal proliferation 24 h after MI. Cardiac catheterization and tissue analysis showed superior cardiac functions, beneficial remodeling and increased capillary density 6 weeks after MI. Concomitant fluorescence-activated cell sorting, co-cultures with neonatal cardiomyocytes, angiogenesis assays, ELISA, western blotting and RAMAN spectroscopy demonstrated that EPO could promote cardiomyogenic differentiation that was specific of tissue origin and enhance paracrine angiogenetic activity in cardiac CD45-CD44+DDR2+ MSCs. Epicardial EPO delivery might be the optimal route for efficient upregulation of regenerative key signals after acute MI. Early EPO-mediated stimulation of mesenchymal proliferation, synergistic angiogenesis with cardiac MSCs and direct induction of TGF-β/WNT signaling in intramyocardial cardiac MSCs could initiate an accelerated healing process that enhances cardiac recovery.
Collapse
Affiliation(s)
- Christian Klopsch
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, 18055 Rostock, Germany .,Department of Cardiac Surgery, Heart Center Rostock, University of Rostock, 18055 Rostock, Germany
| | - Anna Skorska
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, 18055 Rostock, Germany.,Department of Cardiac Surgery, Heart Center Rostock, University of Rostock, 18055 Rostock, Germany
| | - Marion Ludwig
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, 18055 Rostock, Germany.,Department of Cardiac Surgery, Heart Center Rostock, University of Rostock, 18055 Rostock, Germany
| | - Heiko Lemcke
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, 18055 Rostock, Germany.,Department of Cardiac Surgery, Heart Center Rostock, University of Rostock, 18055 Rostock, Germany
| | - Gabriela Maass
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, 18055 Rostock, Germany.,Department of Cardiac Surgery, Heart Center Rostock, University of Rostock, 18055 Rostock, Germany
| | - Ralf Gaebel
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, 18055 Rostock, Germany.,Department of Cardiac Surgery, Heart Center Rostock, University of Rostock, 18055 Rostock, Germany
| | - Martin Beyer
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, 18055 Rostock, Germany.,Department of Cardiac Surgery, Heart Center Rostock, University of Rostock, 18055 Rostock, Germany
| | - Cornelia Lux
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, 18055 Rostock, Germany.,Department of Cardiac Surgery, Heart Center Rostock, University of Rostock, 18055 Rostock, Germany
| | - Anita Toelk
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, 18055 Rostock, Germany.,Department of Cardiac Surgery, Heart Center Rostock, University of Rostock, 18055 Rostock, Germany
| | - Karina Müller
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, 18055 Rostock, Germany.,Department of Cardiac Surgery, Heart Center Rostock, University of Rostock, 18055 Rostock, Germany
| | - Christian Maschmeier
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, 18055 Rostock, Germany.,Department of Cardiac Surgery, Heart Center Rostock, University of Rostock, 18055 Rostock, Germany
| | - Sarah Rohde
- Division of Gastroenterology, Department of Medicine II, Rostock University Medical Center, 18055 Rostock, Germany
| | - Petra Mela
- Department of Tissue Engineering and Textile Implants, AME-Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074 Aachen, Germany
| | - Brigitte Müller-Hilke
- Institute of Immunology & Core Facility for Cell Sorting and Cell Analysis, Rostock University Medical Center, 18055 Rostock, Germany
| | - Stefan Jockenhoevel
- Department of Tissue Engineering and Textile Implants, AME-Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074 Aachen, Germany
| | - Brigitte Vollmar
- Institute for Experimental Surgery, Rostock University Medical Center, 18055 Rostock, Germany
| | - Robert Jaster
- Division of Gastroenterology, Department of Medicine II, Rostock University Medical Center, 18055 Rostock, Germany
| | - Robert David
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, 18055 Rostock, Germany.,Department of Cardiac Surgery, Heart Center Rostock, University of Rostock, 18055 Rostock, Germany
| | - Gustav Steinhoff
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, 18055 Rostock, Germany.,Department of Cardiac Surgery, Heart Center Rostock, University of Rostock, 18055 Rostock, Germany
| |
Collapse
|
14
|
Klopsch C, Skorska A, Ludwig M, Gaebel R, Lemcke H, Kleiner G, Beyer M, Vollmar B, David R, Steinhoff G. Cardiac Mesenchymal Stem Cells Proliferate Early in the Ischemic Heart. Eur Surg Res 2017; 58:341-353. [PMID: 29073604 DOI: 10.1159/000480730] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/28/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND/PURPOSE Cardiac mesenchymal stem cells (MSCs) could stimulate cell-specific regenerative mechanisms after myocardial infarction (MI) depending on spatial origin, distribution, and niche regulation. We aimed at identifying and isolating tissue-specific cardiac MSCs that could contribute to regeneration. METHODS Following permanent ligation of the left anterior descending coronary artery in rats (n = 16), early cardiac tissues and cardiac mononuclear cells (MNCs) were analyzed by immunohistology, confocal laser scanning microscopy, and flow cytometry, respectively. Early postischemic specific MSCs were purified by fluorescence-activated cell sorting, cultivated under standardized culture conditions, and tested for multipotent differentiation in functional identification kits. RESULTS Cardiac MSC niches were detected intramyocardially in cell clusters after MI and characterized by positive expression for vimentin, CD29, CD44, CD90, CD105, PDGFRα, and DDR2. Following myocardial ischemia, proliferation was induced early and proliferation density was approximately 11% in intramyocardial MSC clusters of the peri-infarction border zone. Cluster sizes increased by 157 and 64% in the peri-infarction and noninfarcted areas of infarcted hearts compared with noninfarcted hearts 24 h following MI, respectively. Coincidentally, flow cytometry analyses illustrated postischemic moderate enrichments of CD45-CD44+ and CD45-DDR2+ cardiac MNCs. We enabled isolation of early postischemic culturable cardiac CD45-CD44+DDR2+ MSCs that demonstrated typical clonogenicity with colony-forming unit-fibroblast formation as well as adipogenic, chondrogenic, and osteogenic differentiation. CONCLUSIONS MI triggered early proliferation in specific cardiac MSC niches that were organized in intramyocardial clusters. Following targeted isolation, early postischemic cardiac CD45-CD44+DDR2+ MSCs exhibited typical characteristics with multipotent differentiation capacity and clonogenic expansion.
Collapse
Affiliation(s)
- Christian Klopsch
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, Rostock, Germany.,Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany
| | - Anna Skorska
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, Rostock, Germany.,Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany
| | - Marion Ludwig
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, Rostock, Germany.,Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany
| | - Ralf Gaebel
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, Rostock, Germany.,Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany
| | - Heiko Lemcke
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, Rostock, Germany.,Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany
| | - Gabriela Kleiner
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, Rostock, Germany.,Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany
| | - Martin Beyer
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, Rostock, Germany.,Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany
| | - Brigitte Vollmar
- Institute of Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| | - Robert David
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, Rostock, Germany.,Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany.,Department Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Gustav Steinhoff
- Reference and Translation Center for Cardiac Stem Cell Therapy, Rostock University Medical Center, Rostock, Germany.,Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany.,Department Life, Light and Matter, University of Rostock, Rostock, Germany
| |
Collapse
|
15
|
Pleasant-Jenkins D, Reese C, Chinnakkannu P, Kasiganesan H, Tourkina E, Hoffman S, Kuppuswamy D. Reversal of maladaptive fibrosis and compromised ventricular function in the pressure overloaded heart by a caveolin-1 surrogate peptide. J Transl Med 2017; 97:370-382. [PMID: 28112757 PMCID: PMC5909408 DOI: 10.1038/labinvest.2016.153] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/21/2016] [Accepted: 12/01/2016] [Indexed: 12/19/2022] Open
Abstract
Chronic ventricular pressure overload (PO) results in congestive heart failure (CHF) in which myocardial fibrosis develops in concert with ventricular dysfunction. Caveolin-1 is important in fibrosis in various tissues due to its decreased expression in fibroblasts and monocytes. The profibrotic effects of low caveolin-1 can be blocked with the caveolin-1 scaffolding domain peptide (CSD, a caveolin-1 surrogate) using both mouse models and human cells. We have studied the beneficial effects of CSD on mice in which PO was induced by trans-aortic constriction (TAC). Beneficial effects observed in TAC mice receiving CSD injections daily included: improved ventricular function (increased ejection fraction, stroke volume, and cardiac output; reduced wall thickness); decreased collagen I, collagen chaperone HSP47, fibronectin, and CTGF levels; decreased activation of non-receptor tyrosine kinases Pyk2 and Src; and decreased activation of eNOS. To determine the source of cells that contribute to fibrosis in CHF, flow cytometric studies were performed that suggested that myofibroblasts in the heart are in large part bone marrow-derived. Two CD45+ cell populations were observed. One (Zone 1) contained CD45+/HSP47-/macrophage marker+ cells (macrophages). The second (Zone 2) contained CD45moderate/HSP47+/macrophage marker- cells often defined as fibrocytes. TAC increased the number of cells in Zones 1 and 2 and the level of HSP47 in Zone 2. These studies are a first step in elucidating the mechanism of action of CSD in heart fibrosis and promoting the development of CSD as a novel treatment to reduce fibrosis and improve ventricular function in CHF patients.
Collapse
Affiliation(s)
- Dorea Pleasant-Jenkins
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Charleston, SC, USA
| | - Charles Reese
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | | | - Harinath Kasiganesan
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Charleston, SC, USA
| | - Elena Tourkina
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Stanley Hoffman
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Dhandapani Kuppuswamy
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Charleston, SC, USA
| |
Collapse
|
16
|
Tian J, An X, Niu L. Myocardial fibrosis in congenital and pediatric heart disease. Exp Ther Med 2017; 13:1660-1664. [PMID: 28565750 PMCID: PMC5443200 DOI: 10.3892/etm.2017.4224] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 02/07/2017] [Indexed: 01/13/2023] Open
Abstract
Cardiac fibrosis is a common phenomenon in different types of heart diseases, such as ischemic heart disease, inherited cardiomyopathy mutations, diabetes, and ageing and is associated with morbidity and mortality. Increased accumulation of extracellular matrix (ECM) that impacts cardiac function, is the underlying cause of fibrotic heart disease. There are four different types of cardiac fibrosis, including, reactive interstitial fibrosis, replacement fibrosis, infiltrative interstitial fibrosis and endomyocardial fibrosis. They are involved in the activation and transformation of cardiac fibroblasts to myofibroblasts, which participate in ECM production and fibrotic process and several inflammatory pathways. Besides the ECM proteins, myofibroblasts also express smooth muscle α-actin, SM22 and caldesmon and other markers related to fibrotic process. Most commonly employed techniques to assess myocardial fibrosis include stress echocardiography, cardiac magnetic resonance imaging and positron emission tomography. Because of the involvement of renin-angiotensin-II-aldosterone system, transforming growth factor-β signaling and activin-linked kinase 5 in the mechanisms of cardiac fibrosis, these pathways and the involved proteins are useful as therapeutic targets. However, because of the importance of these pathways in many other physiological functions, their therapeutic targeting needs to be approached with caution.
Collapse
Affiliation(s)
- Jing Tian
- Department of Cardiology, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Xinjiang An
- Department of Cardiology, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Ling Niu
- Department of Cardiology, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
17
|
Abstract
Cardiac fibrosis is a significant global health problem that is closely associated with multiple forms of cardiovascular disease, including myocardial infarction, dilated cardiomyopathy, and diabetes. Fibrosis increases myocardial wall stiffness due to excessive extracellular matrix deposition, causing impaired systolic and diastolic function, and facilitating arrhythmogenesis. As a result, patient morbidity and mortality are often dramatically elevated compared with those with cardiovascular disease but without overt fibrosis, demonstrating that fibrosis itself is both a pathologic response to existing disease and a significant risk factor for exacerbation of the underlying condition. The lack of any specific treatment for cardiac fibrosis in patients suffering from cardiovascular disease is a critical gap in our ability to care for these individuals. Here we provide an overview of the development of cardiac fibrosis, and discuss new research directions that have recently emerged and that may lead to the creation of novel treatments for patients with cardiovascular diseases. Such treatments would, ideally, complement existing therapy by specifically focusing on amelioration of fibrosis.
Collapse
Affiliation(s)
- Danah Al Hattab
- a Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada.,b Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Michael P Czubryt
- a Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada.,b Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| |
Collapse
|
18
|
Lv T, Du Y, Cao N, Zhang S, Gong Y, Bai Y, Wang W, Liu H. Proliferation in cardiac fibroblasts induced by β1-adrenoceptor autoantibody and the underlying mechanisms. Sci Rep 2016; 6:32430. [PMID: 27577254 PMCID: PMC5006240 DOI: 10.1038/srep32430] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 08/09/2016] [Indexed: 12/25/2022] Open
Abstract
Chronic sustained stimulation of β-adrenoceptor is closely related to cardiac fibrosis which is bad for cardiac function. Growing evidence showed that the high prevalence of β1-adrenoceptor autoantibody (β1-AA) in the sera of patients with various types of cardiovascular diseases decreased cardiac function. In the current study, we demonstrated that β1-AA impaired the cardiac function evaluated by echocardiography and that β1-AA triggered cardiac fibrosis in terms of increased expression of α-smooth muscle actin as the marker of myofibroblast and collagen deposition in a passive β1-AA immunized mice model during 16 weeks. Further, we showed that β1-AA activated β1-AR/cAMP/PKA pathway and promoted proliferation in primary cardiac fibroblasts through specific binding to β1-AR but not to β2-AR. Moreover, β1-AA was also likely to promote proliferation in cardiac fibroblasts through activating p38MAPK and ERK1/2 as p38MAPK inhibitor SB203580 and ERK1/2 inhibitor PD98059 partially reversed the proliferative effect. The persistent activating signalling of PKA and P38MAPK in 1 h induced by β1-AA was associated with lacking agonist-induced desensitization phenomena. The conditioned medium from β1-AA-stimulated cardiac fibroblasts induced cardiomyocyte apoptosis, which indicated that β1-AA changed the secretion of cardiac fibroblasts contributing to cardiac injury. These findings will contribute to our understanding of the pathological mechanisms of β1-AA.
Collapse
Affiliation(s)
- Tingting Lv
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China
| | - Yunhui Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Ning Cao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China
| | - Suli Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Yulin Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China
| | - Yan Bai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China
| | - Wen Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| |
Collapse
|
19
|
Stempien-Otero A, Kim DH, Davis J. Molecular networks underlying myofibroblast fate and fibrosis. J Mol Cell Cardiol 2016; 97:153-61. [PMID: 27167848 PMCID: PMC5482716 DOI: 10.1016/j.yjmcc.2016.05.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/02/2016] [Accepted: 05/05/2016] [Indexed: 01/06/2023]
Abstract
Fibrotic remodeling is a hallmark of most forms of cardiovascular disease and a strong prognostic indicator of the advancement towards heart failure. Myofibroblasts, which are a heterogeneous cell-type specialized for extracellular matrix (ECM) secretion and tissue contraction, are the primary effectors of the heart's fibrotic response. This review is focused on defining myofibroblast physiology, its progenitor cell populations, and the core signaling network that orchestrates myofibroblast differentiation as a way of understanding the basic determinants of fibrotic disease in the heart and other tissues.
Collapse
Affiliation(s)
- April Stempien-Otero
- Division of Cardiology, University of Washington School of Medicine, Seattle, WA, USA
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Jennifer Davis
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
20
|
Genetic lineage tracing defines myofibroblast origin and function in the injured heart. Nat Commun 2016; 7:12260. [PMID: 27447449 PMCID: PMC5512625 DOI: 10.1038/ncomms12260] [Citation(s) in RCA: 666] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 06/15/2016] [Indexed: 01/18/2023] Open
Abstract
Cardiac fibroblasts convert to myofibroblasts with injury to mediate healing after acute myocardial infarction (MI) and to mediate long-standing fibrosis with chronic disease. Myofibroblasts remain a poorly defined cell type in terms of their origins and functional effects in vivo. Here we generate Postn (periostin) gene-targeted mice containing a tamoxifen-inducible Cre for cellular lineage-tracing analysis. This Postn allele identifies essentially all myofibroblasts within the heart and multiple other tissues. Lineage tracing with four additional Cre-expressing mouse lines shows that periostin-expressing myofibroblasts in the heart derive from tissue-resident fibroblasts of the Tcf21 lineage, but not endothelial, immune/myeloid or smooth muscle cells. Deletion of periostin+ myofibroblasts reduces collagen production and scar formation after MI. Periostin-traced myofibroblasts also revert back to a less-activated state upon injury resolution. Our results define the myofibroblast as a periostin-expressing cell type necessary for adaptive healing and fibrosis in the heart, which arises from Tcf21+ tissue-resident fibroblasts. The origin and fate of myofibroblasts, the cells responsible for cardiac remodelling and fibrosis, is controversial. Here the authors show that cardiac myofibroblasts express periostin, derive exclusively from tissue-resident fibroblasts, are necessary for scar formation after injury, and can revert back to a less-activated state upon injury resolution.
Collapse
|
21
|
Novel therapeutic strategies targeting fibroblasts and fibrosis in heart disease. Nat Rev Drug Discov 2016; 15:620-638. [PMID: 27339799 DOI: 10.1038/nrd.2016.89] [Citation(s) in RCA: 236] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Our understanding of the functions of cardiac fibroblasts has moved beyond their roles in heart structure and extracellular matrix generation and now includes their contributions to paracrine, mechanical and electrical signalling during ontogenesis and normal cardiac activity. Fibroblasts also have central roles in pathogenic remodelling during myocardial ischaemia, hypertension and heart failure. As key contributors to scar formation, they are crucial for tissue repair after interventions including surgery and ablation. Novel experimental approaches targeting cardiac fibroblasts are promising potential therapies for heart disease. Indeed, several existing drugs act, at least partially, through effects on cardiac connective tissue. This Review outlines the origins and roles of fibroblasts in cardiac development, homeostasis and disease; illustrates the involvement of fibroblasts in current and emerging clinical interventions; and identifies future targets for research and development.
Collapse
|
22
|
Ongstad EL, Gourdie RG. Can heart function lost to disease be regenerated by therapeutic targeting of cardiac scar tissue? Semin Cell Dev Biol 2016; 58:41-54. [PMID: 27234380 DOI: 10.1016/j.semcdb.2016.05.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/18/2016] [Accepted: 05/23/2016] [Indexed: 01/14/2023]
Abstract
Myocardial infarction results in scar tissue that cannot actively contribute to heart mechanical function and frequently causes lethal arrhythmias. The healing response after infarction involves inflammation, biochemical signaling, changes in cellular phenotype, activity, and organization, and alterations in electrical conduction due to variations in cell and tissue geometry and alterations in protein expression, organization, and function - particularly in membrane channels. The intensive research focus on regeneration of myocardial tissues has, as of yet, only met with modest success, with no near-term prospect of improving standard-of-care for patients with heart disease. An alternative concept for novel therapeutic approach is the rejuvenation of cardiac electrical and mechanical properties through the modification of scar tissue. Several peptide therapeutics, locally applied genetic therapies, or delivery of genetically modified cells have shown promise in improving the characteristics of the fibrous scar and post-myocardial infarction prognosis in experimental models. This review highlights several factors that contribute to arrhythmogenesis in scar formation and how these might be targeted to regenerate some of the electrical and mechanical function of the post-MI scar.
Collapse
Affiliation(s)
- Emily L Ongstad
- Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion Research Institute, 2 Riverside Circle, Roanoke, VA 24016, USA.
| | - Robert G Gourdie
- Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion Research Institute, 2 Riverside Circle, Roanoke, VA 24016, USA; Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, 317 Kelly Hall, Stanger Street, Blacksburg, VA 24061, USA; Department of Emergency Medicine, Carilion Clinic, 1906 Belleview Avenue, Roanoke VA 24014, USA.
| |
Collapse
|
23
|
Telocytes in Cardiac Tissue Architecture and Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 913:127-137. [PMID: 27796884 DOI: 10.1007/978-981-10-1061-3_8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
24
|
The Janus face of myofibroblasts in the remodeling heart. J Mol Cell Cardiol 2015; 91:35-41. [PMID: 26690324 DOI: 10.1016/j.yjmcc.2015.11.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/12/2015] [Accepted: 11/14/2015] [Indexed: 01/14/2023]
|
25
|
Fountoulaki K, Dagres N, Iliodromitis EK. Cellular Communications in the Heart. Card Fail Rev 2015; 1:64-68. [PMID: 28785434 PMCID: PMC5490974 DOI: 10.15420/cfr.2015.1.2.64] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/12/2015] [Indexed: 11/04/2022] Open
Abstract
Heart failure is one of the leading causes of morbidity and mortality worldwide. Cardiac remodelling is first an adaptive, becoming a maladaptive, compensatory mechanism that finally causes ventricular dysfunction independently of the etiology of the initial insult. In the present article the authors describe the elements of the human heart, examining their basic functions and their inter-communication under both normal and pathological circumstances. Cardiac myocytes carry out mechanical and electrical functions of the heart and cardiac fibroblasts maintain its structural integrity. Several factors can affect fibroblast activation and under pathological stress they transdifferentiate into myofibroblasts. Endothelial cells have complex biological functions, including the control of vascular permeability, vasomotion, regulation of haemostasis, immune responses and angiogenesis. The extracellular matrix is a complex architectural network consisting of a variety of proteins. Various routes using a plethora of products and mediators contribute to the cross-talk of the myocytes with endothelial cells, extracellular matrix and cardiac fibroblasts. A better understanding of the entire mechanism of cellular communication by the established or the more recently discovered agents will certainly emerge promising new perspectives when looking at the prevention of heart failure and leading to more substantial therapeutic interventions.
Collapse
Affiliation(s)
- Katerina Fountoulaki
- Cardiothoracic Intensive Care Unit, Onassis Cardiac Surgery Centre, Athens, Greece
| | - Nikolaos Dagres
- Second University Department of Cardiology, Attikon General Hospial, University of Athens, Athens, Greece
| | - Efstathios K Iliodromitis
- Second University Department of Cardiology, Attikon General Hospial, University of Athens, Athens, Greece
| |
Collapse
|
26
|
Abstract
Myocardial remodeling following myocardial infarction (MI) is emerging as key causes of chronic infarct mortality. Interleukin-6 is a classic pro-inflammatory cytokine needed to mount an effective immune response. It seems that interleukin-6 acts as an important role in the dynamic and superbly orchestrated process of innate immunity after MI. Interleukin-6 timely suppresses of innate immune signals to prevent the catastrophic consequences of uncontrolled inflammation on cardiac geometry and function, and thus tunes myocardial remodeling. A comprehensive understanding of biological processes of interleukin-6 in innate immunity leading to inflammatory response and disease-related ventricular remodeling is helpful to find the solution of chronic heart failure. To accomplish this, we reviewed the articles of interleukin-6 regard to inflammation, innate immunity, and cardiac remodeling. This review focuses on the role of interleukin-6 that dominates cell-mediated immunity, especially on neutrophils, monocytes, macrophages, and fibroblasts. In addition, we will also briefly discuss other inflammatory cytokines involved in this process within the paper.
Collapse
|
27
|
Dmitrieva RI, Revittser AV, Klukina MA, Sviryaev YV, Korostovtseva LS, Kostareva AA, Zaritskey AY, Shlyakhto EV. Functional properties of bone marrow derived multipotent mesenchymal stromal cells are altered in heart failure patients, and could be corrected by adjustment of expansion strategies. Aging (Albany NY) 2015; 7:14-25. [PMID: 25606985 PMCID: PMC4350322 DOI: 10.18632/aging.100716] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Bone marrow multipotent mesenchymal stromal cells (BM-MMSC) considered as a prospective substrate for cell therapy applications, however adult stem cells could be affected by donor-specific factors: age, gender, medical history. Our aim was to investigate how HF affects the functional properties of BM-MMSC. MATERIALS AND METHODS BM-MMSC from 10 healthy donors (HD), and 16 donors with chronic HF were evaluated for proliferative activity, ability to differentiate, replicative senescence, expression of genes that affect regeneration and fibrosis. The effect of culturing conditions on efficiency of BM-MMSC expansion was determined. RESULTS HF-derived BM-MMSC demonstrated early decrease of proliferative activity and upregulation of genes that control both, regeneration and fibrosis: Tgf-β pathway, synthesis of ECM, remodeling enzymes, adhesion molecules. We assume that these effects were related to increase of frequency of myofibroblast-like CD146+/SMAα+ CFU-F in HF samples; (ii) low seeding density and hypoxia resulted in predominant purification and expansion of CD146+/SMAα- CFU-Fs. (iii) the activity of NPs system was downregulated in HF BM-MMSC; CONCLUSIONS downregulation of NP signaling in combination with upregulation of Tgf-β pathway in BM-MMSC would result in pro-fibrotic phenotype and make these cells non-effective for therapeutic applications; the corrections in culturing strategy resulted in 2(3)-2(7) increase of expansion efficiency.
Collapse
Affiliation(s)
| | - Alla V Revittser
- Federal Almazov Medical Research Centre, St. Petersburg, Russia.,St. Petersburg State Polytechnical University, Branch of Medical Physics and Bioengineering, Russia
| | - Maria A Klukina
- Federal Almazov Medical Research Centre, St. Petersburg, Russia
| | - Yuri V Sviryaev
- Federal Almazov Medical Research Centre, St. Petersburg, Russia
| | | | | | | | | |
Collapse
|
28
|
Epelman S, Liu PP, Mann DL. Role of innate and adaptive immune mechanisms in cardiac injury and repair. Nat Rev Immunol 2015; 15:117-29. [PMID: 25614321 DOI: 10.1038/nri3800] [Citation(s) in RCA: 476] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite the advances that have been made in developing new therapeutics, cardiovascular disease remains the leading cause of worldwide mortality. Therefore, understanding the mechanisms underlying cardiovascular tissue injury and repair is of prime importance. Following cardiac tissue injury, the immune system has an important and complex role in driving both the acute inflammatory response and the regenerative response. This Review summarizes the role of the immune system in cardiovascular disease - focusing on the idea that the immune system evolved to promote tissue homeostasis following injury and/or infection, and that the inherent cost of this evolutionary development is unwanted inflammatory damage.
Collapse
Affiliation(s)
- Slava Epelman
- Toronto Medical Discovery Tower, 101 College Street, TMDT 3903 Toronto, Ontario, M5G 1L7, Canada
| | - Peter P Liu
- University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario, K1Y 4W7, Canada
| | - Douglas L Mann
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
29
|
Tao H, Yang JJ, Shi KH, Deng ZY, Li J. DNA methylation in cardiac fibrosis: new advances and perspectives. Toxicology 2014; 323:125-9. [PMID: 25017140 DOI: 10.1016/j.tox.2014.07.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 07/08/2014] [Accepted: 07/09/2014] [Indexed: 01/22/2023]
Abstract
Cardiac fibrosis is characterized by net accumulation of extracellular matrix (ECM) proteins in the cardiac interstitium, and contributes to both systolic and diastolic dysfunction in many cardiac pathophysiologic conditions. More specifically, cardiac fibroblasts are activated by a variety of pathological stimuli, thereby undergoing proliferation, differentiation to myofibroblasts, and production of various cytokines and ECM proteins. Thus, understanding the biological processes of cardiac fibroblasts will provide novel insights into the underlying mechanisms of cardiac fibrosis. DNA methylation is an important epigenetic mechanism, which often occurs in response to environmental stimuli and is crucial in regulating gene expression. The aberrant methylation of CpG island promoters of selected genes is the prominent epigenetic mechanism by which gene transcription can be effectively silenced. Aberrant hypermethylation of a few selected genes such as RASSF1A plays an important role in facilitating fibrotic fibroblast activation and in driving fibrosis. In this review we will discuss the mechanisms of DNA methylation and their implications for cardiac fibroblasts activation and fibrosis. Control of DNA methylation may serve as a new strategy for anti-fibrotic therapy.
Collapse
Affiliation(s)
- Hui Tao
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei 230601, China; Cardiovascular Research Center, Anhui Medical University, Hefei 230601, China
| | - Jing-Jing Yang
- Department of Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Kai-Hu Shi
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei 230601, China; Cardiovascular Research Center, Anhui Medical University, Hefei 230601, China.
| | - Zi-Yu Deng
- Department of Scientific and Educational, The Second Hospital of Anhui Medical University, Hefei 230032, China.
| | - Jun Li
- School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
30
|
Goldsmith EC, Bradshaw AD, Zile MR, Spinale FG. Myocardial fibroblast-matrix interactions and potential therapeutic targets. J Mol Cell Cardiol 2014; 70:92-9. [PMID: 24472826 PMCID: PMC4005609 DOI: 10.1016/j.yjmcc.2014.01.008] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 01/18/2014] [Accepted: 01/20/2014] [Indexed: 01/18/2023]
Abstract
The cardiac extracellular matrix (ECM) is a dynamic structure, adapting to physiological and pathological stresses placed on the myocardium. Deposition and organization of the matrix fall under the purview of cardiac fibroblasts. While often overlooked compared to myocytes, fibroblasts play a critical role in maintaining ECM homeostasis under normal conditions and in response to pathological stimuli assume an activated, myofibroblast phenotype associated with excessive collagen accumulation contributing to impaired cardiac function. Complete appreciation of fibroblast function is hampered by the lack of fibroblast-specific reagents and the heterogeneity of fibroblast precursors. This is further complicated by our ability to dissect the role of myofibroblasts versus fibroblasts in myocardial in remodeling. This review highlights critical points in the regulation of collagen deposition by fibroblasts, the current panel of molecular tools used to identify fibroblasts and the role of fibroblast-matrix interactions in fibroblast function and differentiation into the myofibroblast phenotype. The clinical potential of exploiting differences between fibroblasts and myofibroblasts and using them to target specific fibroblast populations is also discussed. This article is part of a Special Issue entitled "Myocyte-Fibroblast Signalling in Myocardium."
Collapse
Affiliation(s)
- Edie C Goldsmith
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, USA.
| | - Amy D Bradshaw
- Ralph H. Johnson Department of Veteran's Affairs Medical Center, Charleston, SC, USA; Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, SC, USA
| | - Michael R Zile
- Ralph H. Johnson Department of Veteran's Affairs Medical Center, Charleston, SC, USA; Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, SC, USA
| | - Francis G Spinale
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, USA; Cardiovascular Translational Research Center, University of South Carolina School of Medicine, USA; WJB Dorn Veteran Affairs Medical Center, Columbia, SC, USA
| |
Collapse
|
31
|
Bani D, Nistri S. New insights into the morphogenic role of stromal cells and their relevance for regenerative medicine. lessons from the heart. J Cell Mol Med 2014; 18:363-70. [PMID: 24533677 PMCID: PMC3955144 DOI: 10.1111/jcmm.12247] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 01/14/2014] [Indexed: 02/06/2023] Open
Abstract
The term stromal cells is referred to cells of direct or indirect (hematopoietic) mesenchymal origin, and encompasses different cell populations residing in the connective tissue, which share the ability to produce the macromolecular components of the extracellular matrix and to organize them in the correct spatial assembly. In physiological conditions, stromal cells are provided with the unique ability to shape a proper three-dimensional scaffold and stimulate the growth and differentiation of parenchymal precursors to give rise to tissues and organs. Thus, stromal cells have an essential function in the regulation of organ morphogenesis and regeneration. In pathological conditions, under the influence of local pro-inflammatory mediators, stromal cells can be prompted to differentiate into myofibroblasts, which rather express a fibrogenic phenotype required for prompt deposition of reparatory scar tissue. Indeed, scarring may be interpreted as an emergency healing response to injury typical of evolved animals, like mammals, conceivably directed to preserve survival at the expense of function. However, under appropriate conditions, the original ability of stromal cells to orchestrate organ regeneration, which is typical of some lower vertebrates and mammalian embryos, can be resumed. These concepts underline the importance of expanding the knowledge on the biological properties of stromal cells and their role as key regulators of the three-dimensional architecture of the organs in view of the refinement of the therapeutic protocols of regenerative medicine.
Collapse
Affiliation(s)
- Daniele Bani
- Department of Experimental & Clinical Medicine, Section of Anatomy & Histology, Research Unit of Histology & Embryology, University of Florence, Florence, Italy
| | | |
Collapse
|
32
|
Lajiness JD, Conway SJ. Origin, development, and differentiation of cardiac fibroblasts. J Mol Cell Cardiol 2013; 70:2-8. [PMID: 24231799 DOI: 10.1016/j.yjmcc.2013.11.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 10/23/2013] [Accepted: 11/04/2013] [Indexed: 01/14/2023]
Abstract
Cardiac fibroblasts are the most abundant cell in the mammalian heart. While they have been historically underappreciated in terms of their functional contributions to cardiac development and physiology, they and their activated form, myofibroblasts, are now known to play key roles in both development and disease through structural, paracrine, and electrical interactions with cardiomyocytes. The lack of specific markers for fibroblasts currently convolutes the study of this dynamic cell lineage, but advances in marker analysis and lineage mapping technologies are continuously being made. Understanding how to best utilize these tools, both individually and in combination, will help to elucidate the functional significance of fibroblast-cardiomyocyte interactions in vivo. Here we review what is currently known about the diverse roles played by cardiac fibroblasts and myofibroblasts throughout development and periods of injury with the intent of emphasizing the duality of their nature. This article is part of a Special Issue entitled "Myocyte-Fibroblast Signalling in Myocardium ".
Collapse
Affiliation(s)
- Jacquelyn D Lajiness
- Developmental Biology and Neonatal Medicine Program, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Simon J Conway
- Developmental Biology and Neonatal Medicine Program, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
33
|
Myofibroblasts: trust your heart and let fate decide. J Mol Cell Cardiol 2013; 70:9-18. [PMID: 24189039 DOI: 10.1016/j.yjmcc.2013.10.019] [Citation(s) in RCA: 256] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 10/18/2013] [Accepted: 10/24/2013] [Indexed: 12/27/2022]
Abstract
Cardiac fibrosis is a substantial problem in managing multiple forms of heart disease. Fibrosis results from an unrestrained tissue repair process orchestrated predominantly by the myofibroblast. These are highly specialized cells characterized by their ability to secrete extracellular matrix (ECM) components and remodel tissue due to their contractile properties. This contractile activity of the myofibroblast is ascribed, in part, to the expression of smooth muscle α-actin (αSMA) and other tension-associated structural genes. Myofibroblasts are a newly generated cell type derived largely from residing mesenchymal cells in response to both mechanical and neurohumoral stimuli. Several cytokines, chemokines, and growth factors are induced in the injured heart, and in conjunction with elevated wall tension, specific signaling pathways and downstream effectors are mobilized to initiate myofibroblast differentiation. Here we will review the cell fates that contribute to the myofibroblast as well as nodal molecular signaling effectors that promote their differentiation and activity. We will discuss canonical versus non-canonical transforming growth factor-β (TGFβ), angiotensin II (AngII), endothelin-1 (ET-1), serum response factor (SRF), transient receptor potential (TRP) channels, mitogen-activated protein kinases (MAPKs) and mechanical signaling pathways that are required for myofibroblast transformation and fibrotic disease. This article is part of a Special Issue entitled "Myocyte-Fibroblast Signalling in Myocardium ".
Collapse
|
34
|
Braitsch CM, Kanisicak O, van Berlo JH, Molkentin JD, Yutzey KE. Differential expression of embryonic epicardial progenitor markers and localization of cardiac fibrosis in adult ischemic injury and hypertensive heart disease. J Mol Cell Cardiol 2013; 65:108-19. [PMID: 24140724 DOI: 10.1016/j.yjmcc.2013.10.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 08/28/2013] [Accepted: 10/09/2013] [Indexed: 11/20/2022]
Abstract
During embryonic heart development, the transcription factors Tcf21, Wt1, and Tbx18 regulate activation and differentiation of epicardium-derived cells, including fibroblast lineages. Expression of these epicardial progenitor factors and localization of cardiac fibrosis were examined in mouse models of cardiovascular disease and in human diseased hearts. Following ischemic injury in mice, epicardial fibrosis is apparent in the thickened layer of subepicardial cells that express Wt1, Tbx18, and Tcf21. Perivascular fibrosis with predominant expression of Tcf21, but not Wt1 or Tbx18, occurs in mouse models of pressure overload or hypertensive heart disease, but not following ischemic injury. Areas of interstitial fibrosis in ischemic and hypertensive hearts actively express Tcf21, Wt1, and Tbx18. In all areas of fibrosis, cells that express epicardial progenitor factors are distinct from CD45-positive immune cells. In human diseased hearts, differential expression of Tcf21, Wt1, and Tbx18 also is detected with epicardial, perivascular, and interstitial fibrosis, indicating conservation of reactivated developmental mechanisms in cardiac fibrosis in mice and humans. Together, these data provide evidence for distinct fibrogenic mechanisms that include Tcf21, separate from Wt1 and Tbx18, in different fibroblast populations in response to specific types of cardiac injury.
Collapse
Affiliation(s)
- Caitlin M Braitsch
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, ML 7020, Cincinnati, OH 45229, USA
| | | | | | | | | |
Collapse
|
35
|
Trial J, Cieslik KA, Haudek SB, Duerrschmid C, Entman ML. Th1/M1 conversion to th2/m2 responses in models of inflammation lacking cell death stimulates maturation of monocyte precursors to fibroblasts. Front Immunol 2013; 4:287. [PMID: 24065967 PMCID: PMC3776235 DOI: 10.3389/fimmu.2013.00287] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 09/03/2013] [Indexed: 12/31/2022] Open
Abstract
We have demonstrated that cardiac fibrosis arises from the differentiation of monocyte-derived fibroblasts. We present here evidence that this process requires sequential Th1 and Th2 induction promoting analogous M1 (classically activated) and M2 (alternatively activated) macrophage polarity. Our models are: (1) mice subjected to daily repetitive ischemia and reperfusion (I/R) without infarction and (2) the in vitro transmigration of human mononuclear leukocytes through human cardiac microvascular endothelium. In the mouse heart, leukocytes entered after I/R in response to monocyte chemoattractant protein-1 (MCP-1), which is the major cytokine induced by this protocol. Monocytes within the heart then differentiated into fibroblasts making collagen while bearing the markers of M2 macrophages. T cells were seen in these hearts as well as in the human heart with cardiomyopathy. In the in vitro model, transmigration of the leukocytes was likewise induced by MCP-1 and some monocytes matured into fibroblasts bearing M2 markers. In this model, the MCP-1 stimulus induced a transient Th1 and M1 response that developed into a predominantly Th2 and M2 response. An increase in the Th2 product IL-13 was present in both the human and the mouse models, consistent with its known role in fibrosis. In these simplified models, in which there is no cell death to stimulate an anti-inflammatory response, there is nonetheless a resolution of inflammation enabling a profibrotic environment. This induces the maturation of monocyte precursors into fibroblasts.
Collapse
Affiliation(s)
- Joann Trial
- Division of Cardiovascular Sciences, Department of Medicine, DeBakey Heart Center, Baylor College of Medicine , Houston, TX , USA ; Houston Methodist , Houston, TX , USA
| | | | | | | | | |
Collapse
|
36
|
Cieslik KA, Taffet GE, Crawford JR, Trial J, Mejia Osuna P, Entman ML. AICAR-dependent AMPK activation improves scar formation in the aged heart in a murine model of reperfused myocardial infarction. J Mol Cell Cardiol 2013; 63:26-36. [PMID: 23871790 DOI: 10.1016/j.yjmcc.2013.07.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 07/03/2013] [Accepted: 07/06/2013] [Indexed: 11/19/2022]
Abstract
We have demonstrated that scar formation after myocardial infarction (MI) is associated with an endogenous pool of CD44(pos)CD45(neg) multipotential mesenchymal stem cells (MSC). MSC differentiate into fibroblasts secreting collagen that forms a scar and mature into myofibroblasts that express alpha smooth muscle actin (α-SMA) that stabilizes the scar. In the aging mouse, cardiac repair after MI is associated with impaired differentiation of MSC; MSC derived from the aged hearts form dysfunctional fibroblasts that deposit less collagen in response to transforming growth factor beta-1 (TGF-β1) and poorly mature into myofibroblasts. We found in vitro that the defect in myofibroblast maturation can be remedied by AICAR, which activates non-canonical TGF-β signaling through AMP-activated protein kinase (AMPK). In the present study, we injected aged mice with AICAR and subjected them to 1h occlusion of the left anterior descending artery (LAD) and then reperfusion for up to 30days. AICAR-dependent AMPK signaling led to mobilization of an endogenous CD44(pos)CD45(neg) MSC and its differentiation towards fibroblasts and myofibroblasts in the infarct. This was accompanied by enhanced collagen deposition and collagen fiber maturation in the scar. The AICAR-treated group has demonstrated reduced adverse remodeling as indicated by improved apical end diastolic dimension but no changes in ejection fraction and cardiac output were observed. We concluded that these data indicate the novel, previously not described role of AMPK in the post-MI scar formation. These findings can potentially lead to a new therapeutic strategy for prevention of adverse remodeling in the aging heart.
Collapse
Affiliation(s)
- Katarzyna A Cieslik
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, and The Methodist Hospital, Houston, TX 77030, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Turner NA, Porter KE. Function and fate of myofibroblasts after myocardial infarction. FIBROGENESIS & TISSUE REPAIR 2013; 6:5. [PMID: 23448358 PMCID: PMC3599637 DOI: 10.1186/1755-1536-6-5] [Citation(s) in RCA: 133] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 01/10/2013] [Indexed: 01/27/2023]
Abstract
The importance of cardiac fibroblasts in the regulation of myocardial remodelling following myocardial infarction (MI) is becoming increasingly recognised. Studies over the last few decades have reinforced the concept that cardiac fibroblasts are much more than simple homeostatic regulators of extracellular matrix turnover, but are integrally involved in all aspects of the repair and remodelling of the heart that occurs following MI. The plasticity of fibroblasts is due in part to their ability to undergo differentiation into myofibroblasts. Myofibroblasts are specialised cells that possess a more contractile and synthetic phenotype than fibroblasts, enabling them to effectively repair and remodel the cardiac interstitium to manage the local devastation caused by MI. However, in addition to their key role in cardiac restoration and healing, persistence of myofibroblast activation can drive pathological fibrosis, resulting in arrhythmias, myocardial stiffness and progression to heart failure. The aim of this review is to give an appreciation of both the beneficial and detrimental roles of the myofibroblast in the remodelling heart, to describe some of the major regulatory mechanisms controlling myofibroblast differentiation including recent advances in the microRNA field, and to consider how this cell type could be exploited therapeutically.
Collapse
Affiliation(s)
- Neil A Turner
- Division of Cardiovascular and Diabetes Research, and Multidisciplinary Cardiovascular Research Centre, School of Medicine, University of Leeds, Leeds LS2 9JT, UK.
| | | |
Collapse
|
38
|
Goldsmith EC, Bradshaw AD, Spinale FG. Cellular mechanisms of tissue fibrosis. 2. Contributory pathways leading to myocardial fibrosis: moving beyond collagen expression. Am J Physiol Cell Physiol 2012; 304:C393-402. [PMID: 23174564 DOI: 10.1152/ajpcell.00347.2012] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
While the term "fibrosis" can be misleading in terms of the complex patterns and processes of myocardial extracellular matrix (ECM) remodeling, fibrillar collagen accumulation is a common consequence of relevant pathophysiological stimuli, such as pressure overload (PO) and myocardial infarction (MI). Fibrillar collagen accumulation in both PO and MI is predicated on a number of diverse cellular and extracellular events, which include changes in fibroblast phenotype (transdifferentiation), posttranslational processing and assembly, and finally, degradation. The expansion of a population of transformed fibroblasts/myofibroblasts is a significant cellular event with respect to ECM remodeling in both PO and MI. The concept that this cellular expansion within the myocardial ECM may be due, at least in part, to endothelial-mesenchymal transformation and thereby not dissimilar to events observed in cancer progression holds intriguing future possibilities. Studies regarding determinants of procollagen processing, such as procollagen C-endopeptidase enhancer (PCOLCE), and collagen assembly, such as the secreted protein acidic and rich in cysteine (SPARC), have identified potential new targets for modifying the fibrotic response in both PO and MI. Finally, the transmembrane matrix metalloproteinases, such as MMP-14, underscore the diversity and complexity of this ECM proteolytic family as this protease can degrade the ECM as well as induce a profibrotic response. The growing recognition that the myocardial ECM is a dynamic entity containing a diversity of matricellular and nonstructural proteins as well as proteases and that the fibrillar collagens can change in structure and content in a rapid temporal fashion has opened up new avenues for modulating what was once considered an irreversible event--myocardial fibrosis.
Collapse
Affiliation(s)
- Edie C Goldsmith
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina 29208, USA
| | | | | |
Collapse
|
39
|
Common threads in cardiac fibrosis, infarct scar formation, and wound healing. FIBROGENESIS & TISSUE REPAIR 2012; 5:19. [PMID: 23114500 PMCID: PMC3534582 DOI: 10.1186/1755-1536-5-19] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 10/04/2012] [Indexed: 12/19/2022]
Abstract
Wound healing, cardiac fibrosis, and infarct scar development, while possessing distinct features, share a number of key functional similarities, including extracellular matrix synthesis and remodeling by fibroblasts and myofibroblasts. Understanding the underlying mechanisms that are common to these processes may suggest novel therapeutic approaches for pathologic situations such as fibrosis, or defective wound healing such as hypertrophic scarring or keloid formation. This manuscript will briefly review the major steps of wound healing, and will contrast this process with how cardiac infarct scar formation or interstitial fibrosis occurs. The feasibility of targeting common pro-fibrotic growth factor signaling pathways will be discussed. Finally, the potential exploitation of novel regulators of wound healing and fibrosis (ski and scleraxis), will be examined.
Collapse
|