1
|
Huang L. The role of IL-17 family cytokines in cardiac fibrosis. Front Cardiovasc Med 2024; 11:1470362. [PMID: 39502194 PMCID: PMC11534612 DOI: 10.3389/fcvm.2024.1470362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/29/2024] [Indexed: 11/08/2024] Open
Abstract
Myocardial fibrosis is a common pathological feature in various cardiovascular diseases including myocardial infarction, heart failure, and myocarditis. Generally, persistent myocardial fibrosis correlates with poor prognosis and ranks among the leading causes of death globally. Currently, there is no effective treatment for myocardial fibrosis, partly due to its unclear pathogenic mechanism. Increasing studies have shown IL-17 family cytokines are strongly associated with the initiation and propagation of myocardial fibrosis. This review summarizes the expression, action, and signal transduction mechanisms of IL-17, focusing on its role in fibrosis associated with cardiovascular diseases such as myocardial infarction, heart failure, hypertension, diabetes, and myocarditis. It also discusses its potential as a therapeutic target, offering new insights for the clinical treatment of myocardial fibrosis.
Collapse
Affiliation(s)
- Liqing Huang
- Three Gorges University Hospital of Traditional Chinese Medicine & Yichang Hospital of Traditional Chinese Medicine, Yichang, China
| |
Collapse
|
2
|
Wang M, Mo D, Zhang N, Yu H. Ferroptosis in diabetic cardiomyopathy: Advances in cardiac fibroblast-cardiomyocyte interactions. Heliyon 2024; 10:e35219. [PMID: 39165946 PMCID: PMC11334834 DOI: 10.1016/j.heliyon.2024.e35219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/19/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a common complication of diabetes, and its pathogenesis remains elusive. Ferroptosis, a process dependent on iron-mediated cell death, plays a crucial role in DCM via disrupted iron metabolism, lipid peroxidation, and weakened antioxidant defenses. Hyperglycemia, oxidative stress, and inflammation may exacerbate ferroptosis in diabetes. This review emphasizes the interaction between cardiac fibroblasts and cardiomyocytes in DCM, influencing ferroptosis occurrence. By exploring ferroptosis modulation for potential therapeutic targets, this article offers a fresh perspective on DCM treatment. The study systematically covers the interplay, mechanisms, and targeted drugs linked to ferroptosis in DCM development.
Collapse
Affiliation(s)
| | | | - Ning Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Haichu Yu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| |
Collapse
|
3
|
Samiminemati A, Aprile D, Siniscalco D, Di Bernardo G. Methods to Investigate the Secretome of Senescent Cells. Methods Protoc 2024; 7:52. [PMID: 39051266 PMCID: PMC11270363 DOI: 10.3390/mps7040052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/27/2024] Open
Abstract
The word "secretome" was first used to describe the proteins that cells secrete under different circumstances; however, recent studies have proven the existence of other molecules such as RNA and chemical compounds in the secretome. The study of secretome has significance for the diagnosis and treatment of disease as it provides insight into cellular functions, including immune responses, development, and homeostasis. By halting cell division, cellular senescence plays a role in both cancer defense and aging by secreting substances known as senescence-associated secretory phenotypes (SASP). A variety of techniques could be used to analyze the secretome: protein-based approaches like mass spectrometry and protein microarrays, nucleic acid-based methods like RNA sequencing, microarrays, and in silico prediction. Each method offers unique advantages and limitations in characterizing secreted molecules. Top-down and bottom-up strategies for thorough secretome analysis are became possible by mass spectrometry. Understanding cellular function, disease causes, and proper treatment targets is aided by these methodologies. Their approaches, benefits, and drawbacks will all be discussed in this review.
Collapse
Affiliation(s)
- Afshin Samiminemati
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Luigi Vanvitelli Campania University, 80138 Naples, Italy; (A.S.); (D.A.); (D.S.)
| | - Domenico Aprile
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Luigi Vanvitelli Campania University, 80138 Naples, Italy; (A.S.); (D.A.); (D.S.)
| | - Dario Siniscalco
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Luigi Vanvitelli Campania University, 80138 Naples, Italy; (A.S.); (D.A.); (D.S.)
| | - Giovanni Di Bernardo
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Luigi Vanvitelli Campania University, 80138 Naples, Italy; (A.S.); (D.A.); (D.S.)
- Sbarro Health Research Organization, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
4
|
Stempien A, Josvai M, Notbohm J, Zhang J, Kamp TJ, Crone WC. Influence of Remodeled ECM and Co-culture with iPSC-Derived Cardiac Fibroblasts on the Mechanical Function of Micropatterned iPSC-Derived Cardiomyocytes. Cardiovasc Eng Technol 2024; 15:264-278. [PMID: 38448643 PMCID: PMC11239313 DOI: 10.1007/s13239-024-00711-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 01/02/2024] [Indexed: 03/08/2024]
Abstract
INTRODUCTION In native heart tissue, functions of cardiac fibroblasts (CFs) include synthesis, remodeling, and degradation of the extracellular matrix (ECM) as well as secreting factors that regulate cardiomyocyte (CM) function. The influence of direct co-culture and CF-derived ECM on CM mechanical function are not fully understood. METHODS Here we use an engineered culture platform that provides control over ECM geometry and substrate stiffness to evaluate the influence of iPSC-CFs, and the ECM they produce, on the mechanical function of iPSC-CMs. Mechanical analysis was performed using digital image correlation to quantify maximum contractile strain, spontaneous contraction rate, and full-field organization of the contractions. RESULTS When cultured alone, iPSC-CFs produce and remodel the ECM into fibers following the underlying 15° chevron patterned ECM. The substrates were decellularized and confirmed to have highly aligned fibers that covered a large fraction of the pattern area before reseeding with iPSC-CMs, alone or in co-culture with iPSC-CFs. When seeded on decellularized ECM, larger maximum contractile strains were observed in the co-culture condition compared to the CM Only condition. No significant difference was found in contractile strain between the Matrigel and decellularized ECM conditions; however, the spontaneous contraction rate was lower in the decellularized ECM condition. A methodology for quantifying alignment of cell contraction across the entire field of view was developed based on trajectories approximating the cell displacements during contraction. Trajectory alignment was unaltered by changes in culture or ECM conditions. CONCLUSIONS These combined observations highlight the important role CFs play in vivo and the need for models that enable a quantitative approach to examine interactions between the CFs and CMs, as well as the interactions of these cells with the ECM.
Collapse
Affiliation(s)
- A Stempien
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
| | - M Josvai
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
| | - J Notbohm
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - J Zhang
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - T J Kamp
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - W C Crone
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA.
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
5
|
Zygmunciak P, Stróżna K, Błażowska O, Mrozikiewicz-Rakowska B. Extracellular Vesicles in Diabetic Cardiomyopathy-State of the Art and Future Perspectives. Int J Mol Sci 2024; 25:6117. [PMID: 38892303 PMCID: PMC11172920 DOI: 10.3390/ijms25116117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/24/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Cardiovascular complications are the most deadly and cost-driving effects of diabetes mellitus (DM). One of them, which is steadily attracting attention among scientists, is diabetes-induced heart failure, also known as diabetic cardiomyopathy (DCM). Despite significant progress in the research concerning the disease, a universally accepted definition is still lacking. The pathophysiology of the processes accelerating heart insufficiency in diabetic patients on molecular and cellular levels also remains elusive. However, the recent interest concerning extracellular vesicles (EVs) has brought promise to further clarifying the pathological events that lead to DCM. In this review, we sum up recent investigations on the involvement of EVs in DCM and show their therapeutic and indicatory potential.
Collapse
Affiliation(s)
| | - Katarzyna Stróżna
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland; (P.Z.)
| | - Olga Błażowska
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland; (P.Z.)
| | - Beata Mrozikiewicz-Rakowska
- Department of Endocrinology, Centre of Postgraduate Medical Education, Marymoncka St. 99/103, 01-813 Warsaw, Poland
| |
Collapse
|
6
|
Zhang H, Dhalla NS. The Role of Pro-Inflammatory Cytokines in the Pathogenesis of Cardiovascular Disease. Int J Mol Sci 2024; 25:1082. [PMID: 38256155 PMCID: PMC10817020 DOI: 10.3390/ijms25021082] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
With cardiovascular disease (CVD) being a primary source of global morbidity and mortality, it is crucial that we understand the molecular pathophysiological mechanisms at play. Recently, numerous pro-inflammatory cytokines have been linked to several different CVDs, which are now often considered an adversely pro-inflammatory state. These cytokines most notably include interleukin-6 (IL-6),tumor necrosis factor (TNF)α, and the interleukin-1 (IL-1) family, amongst others. Not only does inflammation have intricate and complex interactions with pathophysiological processes such as oxidative stress and calcium mishandling, but it also plays a role in the balance between tissue repair and destruction. In this regard, pre-clinical and clinical evidence has clearly demonstrated the involvement and dynamic nature of pro-inflammatory cytokines in many heart conditions; however, the clinical utility of the findings so far remains unclear. Whether these cytokines can serve as markers or risk predictors of disease states or act as potential therapeutic targets, further extensive research is needed to fully understand the complex network of interactions that these molecules encompass in the context of heart disease. This review will highlight the significant advances in our understanding of the contributions of pro-inflammatory cytokines in CVDs, including ischemic heart disease (atherosclerosis, thrombosis, acute myocardial infarction, and ischemia-reperfusion injury), cardiac remodeling (hypertension, cardiac hypertrophy, cardiac fibrosis, cardiac apoptosis, and heart failure), different cardiomyopathies as well as ventricular arrhythmias and atrial fibrillation. In addition, this article is focused on discussing the shortcomings in both pathological and therapeutic aspects of pro-inflammatory cytokines in CVD that still need to be addressed by future studies.
Collapse
Affiliation(s)
- Hannah Zhang
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
7
|
Zhang S, Yang Y, Lv X, Liu W, Zhu S, Wang Y, Xu H. Unraveling the Intricate Roles of Exosomes in Cardiovascular Diseases: A Comprehensive Review of Physiological Significance and Pathological Implications. Int J Mol Sci 2023; 24:15677. [PMID: 37958661 PMCID: PMC10650316 DOI: 10.3390/ijms242115677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/21/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Exosomes, as potent intercellular communication tools, have garnered significant attention due to their unique cargo-carrying capabilities, which enable them to influence diverse physiological and pathological functions. Extensive research has illuminated the biogenesis, secretion, and functions of exosomes. These vesicles are secreted by cells in different states, exerting either protective or harmful biological functions. Emerging evidence highlights their role in cardiovascular disease (CVD) by mediating comprehensive interactions among diverse cell types. This review delves into the significant impacts of exosomes on CVD under stress and disease conditions, including coronary artery disease (CAD), myocardial infarction, heart failure, and other cardiomyopathies. Focusing on the cellular signaling and mechanisms, we explore how exosomes mediate multifaceted interactions, particularly contributing to endothelial dysfunction, oxidative stress, and apoptosis in CVD pathogenesis. Additionally, exosomes show great promise as biomarkers, reflecting differential expressions of NcRNAs (miRNAs, lncRNAs, and circRNAs), and as therapeutic carriers for targeted CVD treatment. However, the specific regulatory mechanisms governing exosomes in CVD remain incomplete, necessitating further exploration of their characteristics and roles in various CVD-related contexts. This comprehensive review aims to provide novel insights into the biological implications of exosomes in CVD and offer innovative perspectives on the diagnosis and treatment of CVD.
Collapse
Affiliation(s)
| | | | | | | | | | - Ying Wang
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China; (S.Z.); (Y.Y.); (X.L.); (W.L.); (S.Z.)
| | - Hongfei Xu
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China; (S.Z.); (Y.Y.); (X.L.); (W.L.); (S.Z.)
| |
Collapse
|
8
|
Nassal DM, Shaheen R, Patel NJ, Yu J, Leahy N, Bibidakis D, Parinandi NL, Hund TJ. Spectrin-Based Regulation of Cardiac Fibroblast Cell-Cell Communication. Cells 2023; 12:748. [PMID: 36899883 PMCID: PMC10001335 DOI: 10.3390/cells12050748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/14/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Cardiac fibroblasts (CFs) maintain the fibrous extracellular matrix (ECM) that supports proper cardiac function. Cardiac injury induces a transition in the activity of CFs to promote cardiac fibrosis. CFs play a critical role in sensing local injury signals and coordinating the organ level response through paracrine communication to distal cells. However, the mechanisms by which CFs engage cell-cell communication networks in response to stress remain unknown. We tested a role for the action-associated cytoskeletal protein βIV-spectrin in regulating CF paracrine signaling. Conditioned culture media (CCM) was collected from WT and βIV-spectrin deficient (qv4J) CFs. WT CFs treated with qv4J CCM showed increased proliferation and collagen gel compaction compared to control. Consistent with the functional measurements, qv4J CCM contained higher levels of pro-inflammatory and pro-fibrotic cytokines and increased concentration of small extracellular vesicles (30-150 nm diameter, exosomes). Treatment of WT CFs with exosomes isolated from qv4J CCM induced a similar phenotypic change as that observed with complete CCM. Treatment of qv4J CFs with an inhibitor of the βIV-spectrin-associated transcription factor, STAT3, decreased the levels of both cytokines and exosomes in conditioned media. This study expands the role of the βIV-spectrin/STAT3 complex in stress-induced regulation of CF paracrine signaling.
Collapse
Affiliation(s)
- Drew M. Nassal
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Rebecca Shaheen
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Nehal J. Patel
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Jane Yu
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Nick Leahy
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Dimitra Bibidakis
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Narasimham L. Parinandi
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Internal Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Thomas J. Hund
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
9
|
Trager LE, Lyons M, Kuznetsov A, Sheffield C, Roh K, Freeman R, Rhee J, Guseh JS, Li H, Rosenzweig A. Beyond cardiomyocytes: Cellular diversity in the heart's response to exercise. JOURNAL OF SPORT AND HEALTH SCIENCE 2022:S2095-2546(22)00125-9. [PMID: 36549585 PMCID: PMC10362490 DOI: 10.1016/j.jshs.2022.12.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/24/2022] [Accepted: 11/07/2022] [Indexed: 06/17/2023]
Abstract
Cardiomyocytes comprise ∼70% to 85% of the total volume of the adult mammalian heart but only about 25% to 35% of its total number of cells. Advances in single cell and single nuclei RNA sequencing have greatly facilitated investigation into and increased appreciation of the potential functions of non-cardiomyocytes in the heart. While much of this work has focused on the relationship between non-cardiomyocytes, disease, and the heart's response to pathological stress, it will also be important to understand the roles that these cells play in the healthy heart, cardiac homeostasis, and the response to physiological stress such as exercise. The present review summarizes recent research highlighting dynamic changes in non-cardiomyocytes in response to the physiological stress of exercise. Of particular interest are changes in fibrotic pathways, the cardiac vasculature, and immune or inflammatory cells. In many instances, limited data are available about how specific lineages change in response to exercise or whether the changes observed are functionally important, underscoring the need for further research.
Collapse
Affiliation(s)
- Lena E Trager
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; University of Minnesota Medical School, Minneapolis, MI 55455, USA
| | - Margaret Lyons
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Alexandra Kuznetsov
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Cedric Sheffield
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Kangsan Roh
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Anesthesiology and Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rebecca Freeman
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - James Rhee
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Anesthesiology and Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - J Sawalla Guseh
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Haobo Li
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Anthony Rosenzweig
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Institute for Heart and Brain Health, University of Michigan Medical Center, Ann Arbor, MI 48109, USA.
| |
Collapse
|
10
|
Wang Y, Bai L, Wen J, Zhang F, Gu S, Wang F, Yin J, Wang N. Cardiac-specific renalase overexpression alleviates CKD-induced pathological cardiac remodeling in mice. Front Cardiovasc Med 2022; 9:1061146. [PMID: 36588579 PMCID: PMC9798007 DOI: 10.3389/fcvm.2022.1061146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Introduction CKD-induced pathological cardiac remodeling is characterized by myocardial hypertrophy and cardiac fibrosis. The available therapeutic options are limited, it is thus urgently needed to identify novel therapeutic targets. Renalase (RNLS) is a newly discovered protein secreted by the kidney and was found beneficial in many renal diseases. But whether it exerts protective effects on cardiac remodeling in CKD remains unclear. Methods RNLS knockout (KO) and wild-type (WT) mice were both used to build CKD models and the adeno-associated virus (AAV9) system was used to overexpress RNLS cardiac specifically. Echocardiography was performed to detect cardiac structural changes every 6 weeks until 18 weeks post-surgery. High throughput sequencing was performed to understand the underlying mechanisms and the effects of RNLS on cardiac fibroblasts were validated in vitro. Results Knockout of RNLS aggravated cardiac remodeling in CKD, while RNLS cardiac-specific overexpression significantly reduced left ventricular hypertrophy and cardiac fibrosis induced by CKD. The following RNA-sequencing analysis revealed that RNLS significantly downregulated the extracellular matrix (ECM) receptor interaction pathway, ECM organization, and several ECM-related proteins. GSEA results showed RNLS significantly downregulated several profibrotic biological processes of cardiac fibroblasts which were upregulated by CKD, including fibroblast proliferation, leukocyte migration, antigen presentation, cytokine production, and epithelial-mesenchymal transition (EMT). In vitro, we validated that RNLS reduced the primary cardiac fibroblast proliferation and α-SMA expression stimulated by TGF-β. Conclusion In this study, we examined the cardioprotective role of RNLS in CKD-induced cardiac remodeling. RNLS may be a potential therapeutic factor that exerts an anti-fibrotic effect in pathological cardiac remodeling.
Collapse
Affiliation(s)
- Yi Wang
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Linnan Bai
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiejun Wen
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangfei Zhang
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sijie Gu
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Wang
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianyong Yin
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,*Correspondence: Jianyong Yin,
| | - Niansong Wang
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Niansong Wang,
| |
Collapse
|
11
|
Duangrat R, Parichatikanond W, Morales NP, Pinthong D, Mangmool S. Sustained AT1R stimulation induces upregulation of growth factors in human cardiac fibroblasts via Gαq/TGF-β/ERK signaling that influences myocyte hypertrophy. Eur J Pharmacol 2022; 937:175384. [DOI: 10.1016/j.ejphar.2022.175384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022]
|
12
|
Braidotti N, Chen SN, Long CS, Cojoc D, Sbaizero O. Piezo1 Channel as a Potential Target for Hindering Cardiac Fibrotic Remodeling. Int J Mol Sci 2022; 23:8065. [PMID: 35897650 PMCID: PMC9330509 DOI: 10.3390/ijms23158065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 02/01/2023] Open
Abstract
Fibrotic tissues share many common features with neoplasms where there is an increased stiffness of the extracellular matrix (ECM). In this review, we present recent discoveries related to the role of the mechanosensitive ion channel Piezo1 in several diseases, especially in regulating tumor progression, and how this can be compared with cardiac mechanobiology. Based on recent findings, Piezo1 could be upregulated in cardiac fibroblasts as a consequence of the mechanical stress and pro-inflammatory stimuli that occurs after myocardial injury, and its increased activity could be responsible for a positive feedback loop that leads to fibrosis progression. The increased Piezo1-mediated calcium flow may play an important role in cytoskeleton reorganization since it induces actin stress fibers formation, a well-known characteristic of fibroblast transdifferentiation into the activated myofibroblast. Moreover, Piezo1 activity stimulates ECM and cytokines production, which in turn promotes the phenoconversion of adjacent fibroblasts into new myofibroblasts, enhancing the invasive character. Thus, by assuming the Piezo1 involvement in the activation of intrinsic fibroblasts, recruitment of new myofibroblasts, and uncontrolled excessive ECM production, a new approach to blocking the fibrotic progression can be predicted. Therefore, targeted therapies against Piezo1 could also be beneficial for cardiac fibrosis.
Collapse
Affiliation(s)
- Nicoletta Braidotti
- Department of Physics, University of Trieste, Via A. Valerio 2, 34127 Trieste, Italy;
- Institute of Materials, National Research Council of Italy (CNR-IOM), Area Science Park Basovizza, Strada Statale 14, Km 163,5, 34149 Trieste, Italy;
| | - Suet Nee Chen
- CU-Cardiovascular Institute, University of Colorado Anschutz Medical Campus, 12700 East 19th Ave., Aurora, CO 80045, USA;
| | - Carlin S. Long
- Center for the Prevention of Heart and Vascular Disease, University of California, 555 Mission Bay Blvd South, Rm 352K, San Francisco, CA 94143, USA;
| | - Dan Cojoc
- Institute of Materials, National Research Council of Italy (CNR-IOM), Area Science Park Basovizza, Strada Statale 14, Km 163,5, 34149 Trieste, Italy;
| | - Orfeo Sbaizero
- Department of Engineering and Architecture, University of Trieste, Via A. Valerio 6/A, 34127 Trieste, Italy
| |
Collapse
|
13
|
Germena G, Zelarayán LC, Hinkel R. Cellular Chitchatting: Exploring the Role of Exosomes as Cardiovascular Risk Factors. Front Cell Dev Biol 2022; 10:860005. [PMID: 35433670 PMCID: PMC9008366 DOI: 10.3389/fcell.2022.860005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/16/2022] [Indexed: 11/24/2022] Open
Abstract
Exosomes are small bi-lipid membranous vesicles (30–150 nm) containing different biological material such as proteins, lipids and nucleic acid. These small vesicles, inducing a cell to cell signaling pathway, are able to mediate multidirectional crosstalk to maintain homeostasis or modulate disease processes. With their various contents, exosomes sort and transfer specific information from their origin to a recipient cell, from a tissue or organ in the close proximity or at distance, generating an intra-inter tissue or organ communication. In the last decade exosomes have been identified in multiple organs and fluids under different pathological conditions. In particular, while the content and the abundance of exosome is now a diagnostic marker for cardiovascular diseases, their role in context-specific physiological and pathophysiological conditions in the cardiovascular system remains largely unknown. We summarize here the current knowledge on the role of exosomes as mediators of cardiovascular diseases in several pathophysiological conditions such as atherosclerosis and diabetes. In addition, we describe evidence of intercellular connection among multiple cell type (cardiac, vasculature, immune cells) as well as the challenge of their in vivo analysis.
Collapse
Affiliation(s)
- Giulia Germena
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- *Correspondence: Giulia Germena, ; Rabea Hinkel,
| | - Laura Cecilia Zelarayán
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
| | - Rabea Hinkel
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour (ITTN), Stiftung Tierärztliche Hochschule Hannover, University of Veterinary Medicine, Hannover, Germany
- *Correspondence: Giulia Germena, ; Rabea Hinkel,
| |
Collapse
|
14
|
Haryono A, Ikeda K, Nugroho DB, Ogata T, Tsuji Y, Matoba S, Moriwaki K, Kitagawa H, Igarashi M, Hirata KI, Emoto N. ChGn-2 Plays a Cardioprotective Role in Heart Failure Caused by Acute Pressure Overload. J Am Heart Assoc 2022; 11:e023401. [PMID: 35322673 PMCID: PMC9075488 DOI: 10.1161/jaha.121.023401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Cardiac extracellular matrix is critically involved in cardiac homeostasis, and accumulation of chondroitin sulfate glycosaminoglycans (CS-GAGs) was previously shown to exacerbate heart failure by augmenting inflammation and fibrosis at the chronic phase. However, the mechanism by which CS-GAGs affect cardiac functions remains unclear, especially at the acute phase. Methods and Results We explored a role of CS-GAG in heart failure using mice with target deletion of ChGn-2 (chondroitin sulfate N-acetylgalactosaminyltransferase-2) that elongates CS chains of glycosaminoglycans. Heart failure was induced by transverse aortic constriction in mice. The role of CS-GAG derived from cardiac fibroblasts in cardiomyocyte death was analyzed. Cardiac fibroblasts were subjected to cyclic mechanical stretch that mimics increased workload in the heart. Significant CS-GAGs accumulation was detected in the heart of wild-type mice after transverse aortic constriction, which was substantially reduced in ChGn-2-/- mice. Loss of ChGn-2 deteriorated the cardiac dysfunction caused by pressure overload, accompanied by augmented cardiac hypertrophy and increased cardiomyocyte apoptosis. Cyclic mechanical stretch increased ChGn-2 expression and enhanced glycosaminoglycan production in cardiac fibroblasts. Conditioned medium derived from the stretched cardiac fibroblasts showed cardioprotective effects, which was abolished by CS-GAGs degradation. We found that CS-GAGs elicits cardioprotective effects via dual pathway; direct pathway through interaction with CD44, and indirect pathway through binding to and activating insulin-like growth factor-1. Conclusions Our data revealed the cardioprotective effects of CS-GAGs; therefore, CS-GAGs may play biphasic role in the development of heart failure; cardioprotective role at acute phase despite its possible unfavorable role in the advanced phase.
Collapse
Affiliation(s)
- Andreas Haryono
- Division of Cardiovascular Medicine Department of Internal Medicine Kobe University Graduate School of Medicine Kobe Japan.,Laboratory of Clinical Pharmaceutical Science Kobe Pharmaceutical University Kobe Japan
| | - Koji Ikeda
- Laboratory of Clinical Pharmaceutical Science Kobe Pharmaceutical University Kobe Japan.,Department of Epidemiology for Longevity and Regional Health Kyoto Prefectural University of Medicine Kyoto Japan.,Department of Cardiology Kyoto Prefectural University of Medicine Kyoto Japan
| | - Dhite Bayu Nugroho
- Department of Internal Medicine Faculty of Medicine, Public Health, and Nursing Gadjah Mada University Indonesia
| | - Takehiro Ogata
- Department of Pathology and Cell Regulation Kyoto Prefectural University of Medicine Kyoto Japan
| | - Yumika Tsuji
- Department of Cardiology Kyoto Prefectural University of Medicine Kyoto Japan
| | - Satoaki Matoba
- Department of Cardiology Kyoto Prefectural University of Medicine Kyoto Japan
| | - Kensuke Moriwaki
- Comprehensive Unit for Health Economic Evidence Review and Decision Support (CHEERS) Research Organization of Science and TechnologyRitsumeikan University Kyoto Japan
| | - Hiroshi Kitagawa
- Laboratory of Biochemistry Kobe Pharmaceutical University Kobe Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology Graduate School of Medical and Dental Sciences and Trans-disciplinary Program Niigata University Niigata Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine Department of Internal Medicine Kobe University Graduate School of Medicine Kobe Japan
| | - Noriaki Emoto
- Division of Cardiovascular Medicine Department of Internal Medicine Kobe University Graduate School of Medicine Kobe Japan.,Laboratory of Clinical Pharmaceutical Science Kobe Pharmaceutical University Kobe Japan
| |
Collapse
|
15
|
Exosomes in cardiovascular diseases: a blessing or a sin for the mankind. Mol Cell Biochem 2022; 477:833-847. [PMID: 35064412 DOI: 10.1007/s11010-021-04328-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022]
Abstract
Cardiovascular diseases (CVDs) comprises disorders of blood vessels and heart. Multiple cells in the heart suggests that hetero-cellular communication, which is an important aspect in heart functioning and there is a need to elucidate the way in which this inter-cellular communication occurs. Now a days, exosomal research has gained much attention. Exosomes, nano-shuttles, are EVs with diameters ranging from 40 to 160 nm (average 100 nm), secreted by body cells. These vesicles act as cell-to-cell communicators and are carriers of important biomolecules such as RNAs, miRNAs, Proteins and lipids. Exosomes can change the gene expression of the recipient cells, thereby, changes the cellular characteristics. Exosomes have known to play an essential role in protection as well as progression of various cardiovascular diseases. In the present review, role of exosomes in various CVDs have been discussed.
Collapse
|
16
|
Onódi Z, Visnovitz T, Kiss B, Hambalkó S, Koncz A, Ágg B, Váradi B, Tóth VÉ, Nagy RN, Gergely TG, Gergő D, Makkos A, Pelyhe C, Varga N, Reé D, Apáti Á, Leszek P, Kovács T, Nagy N, Ferdinandy P, Buzás EI, Görbe A, Giricz Z, Varga ZV. Systematic transcriptomic and phenotypic characterization of human and murine cardiac myocyte cell lines and primary cardiomyocytes reveals serious limitations and low resemblances to adult cardiac phenotype. J Mol Cell Cardiol 2021; 165:19-30. [PMID: 34959166 DOI: 10.1016/j.yjmcc.2021.12.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/19/2021] [Accepted: 12/10/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Cardiac cell lines and primary cells are widely used in cardiovascular research. Despite increasing number of publications using these models, comparative characterization of these cell lines has not been performed, therefore, their limitations are undetermined. We aimed to compare cardiac cell lines to primary cardiomyocytes and to mature cardiac tissues in a systematic manner. METHODS AND RESULTS Cardiac cell lines (H9C2, AC16, HL-1) were differentiated with widely used protocols. Left ventricular tissue, neonatal primary cardiomyocytes, and human induced pluripotent stem cell-derived cardiomyocytes served as reference tissue or cells. RNA expression of cardiac markers (e.g. Tnnt2, Ryr2) was markedly lower in cell lines compared to references. Differentiation induced increase in cardiac- and decrease in embryonic markers however, the overall transcriptomic profile and annotation to relevant biological processes showed consistently less pronounced cardiac phenotype in all cell lines in comparison to the corresponding references. Immunocytochemistry confirmed low expressions of structural protein sarcomeric alpha-actinin, troponin I and caveolin-3 in cell lines. Susceptibility of cell lines to sI/R injury in terms of viability as well as mitochondrial polarization differed from the primary cells irrespective of their degree of differentiation. CONCLUSION Expression patterns of cardiomyocyte markers and whole transcriptomic profile, as well as response to sI/R, and to hypertrophic stimuli indicate low-to-moderate similarity of cell lines to primary cells/cardiac tissues regardless their differentiation. Low resemblance of cell lines to mature adult cardiac tissue limits their potential use. Low translational value should be taken into account while choosing a particular cell line to model cardiomyocytes.
Collapse
Affiliation(s)
- Zsófia Onódi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary; MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Tamás Visnovitz
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Bernadett Kiss
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Szabolcs Hambalkó
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Anna Koncz
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Bence Ágg
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Barnabás Váradi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Viktória É Tóth
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary; MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Regina N Nagy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Tamás G Gergely
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary; MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Dorottya Gergő
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
| | - András Makkos
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Csilla Pelyhe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Nóra Varga
- Research Centre for Natural Sciences, Institute of Enzymology, Budapest, Hungary; ELKH-Research Centre for Natural Sciences, Institute of Enzymology, Budapest, Hungary
| | - Dóra Reé
- Research Centre for Natural Sciences, Institute of Enzymology, Budapest, Hungary; ELKH-Research Centre for Natural Sciences, Institute of Enzymology, Budapest, Hungary
| | - Ágota Apáti
- Research Centre for Natural Sciences, Institute of Enzymology, Budapest, Hungary; ELKH-Research Centre for Natural Sciences, Institute of Enzymology, Budapest, Hungary
| | - Przemyslaw Leszek
- Department of Heart Failure and Transplantology, Cardinal Stefan Wyszyński National Institute of Cardiology, Warszawa, Poland
| | - Tamás Kovács
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Nándor Nagy
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Edit I Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary; HCEMM-SU Extracellular Vesicle Research Group, Hungary; ELKH-SE Immune-Proteogenomics Extracellular Vesicle Research Group, Hungary
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary; MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary.
| |
Collapse
|
17
|
Nicin L, Wagner JUG, Luxán G, Dimmeler S. Fibroblast-mediated intercellular crosstalk in the healthy and diseased heart. FEBS Lett 2021; 596:638-654. [PMID: 34787896 DOI: 10.1002/1873-3468.14234] [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] [Received: 09/22/2021] [Revised: 10/28/2021] [Accepted: 11/04/2021] [Indexed: 01/07/2023]
Abstract
Cardiac fibroblasts constitute a major cell population in the heart. They secrete extracellular matrix components and various other factors shaping the microenvironment of the heart. In silico analysis of intercellular communication based on single-cell RNA sequencing revealed that fibroblasts are the source of the majority of outgoing signals to other cell types. This observation suggests that fibroblasts play key roles in orchestrating cellular interactions that maintain organ homeostasis but that can also contribute to disease states. Here, we will review the current knowledge of fibroblast interactions in the healthy, diseased, and aging heart. We focus on the interactions that fibroblasts establish with other cells of the heart, specifically cardiomyocytes, endothelial cells and immune cells, and particularly those relying on paracrine, electrical, and exosomal communication modes.
Collapse
Affiliation(s)
- Luka Nicin
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany.,German Center for Cardiovascular Research (DZHK), Frankfurt am Main, Germany.,Cardio-Pulmonary Institute (CPI), Frankfurt am Main, Germany
| | - Julian U G Wagner
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany.,German Center for Cardiovascular Research (DZHK), Frankfurt am Main, Germany.,Cardio-Pulmonary Institute (CPI), Frankfurt am Main, Germany
| | - Guillermo Luxán
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany.,German Center for Cardiovascular Research (DZHK), Frankfurt am Main, Germany.,Cardio-Pulmonary Institute (CPI), Frankfurt am Main, Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany.,German Center for Cardiovascular Research (DZHK), Frankfurt am Main, Germany.,Cardio-Pulmonary Institute (CPI), Frankfurt am Main, Germany
| |
Collapse
|
18
|
Martins-Marques T. Connecting different heart diseases through intercellular communication. Biol Open 2021; 10:bio058777. [PMID: 34494646 PMCID: PMC8443862 DOI: 10.1242/bio.058777] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 07/12/2021] [Indexed: 12/22/2022] Open
Abstract
Well-orchestrated intercellular communication networks are pivotal to maintaining cardiac homeostasis and to ensuring adaptative responses and repair after injury. Intracardiac communication is sustained by cell-cell crosstalk, directly via gap junctions (GJ) and tunneling nanotubes (TNT), indirectly through the exchange of soluble factors and extracellular vesicles (EV), and by cell-extracellular matrix (ECM) interactions. GJ-mediated communication between cardiomyocytes and with other cardiac cell types enables electrical impulse propagation, required to sustain synchronized heart beating. In addition, TNT-mediated organelle transfer has been associated with cardioprotection, whilst communication via EV plays diverse pathophysiological roles, being implicated in angiogenesis, inflammation and fibrosis. Connecting various cell populations, the ECM plays important functions not only in maintaining the heart structure, but also acting as a signal transducer for intercellular crosstalk. Although with distinct etiologies and clinical manifestations, intercellular communication derailment has been implicated in several cardiac disorders, including myocardial infarction and hypertrophy, highlighting the importance of a comprehensive and integrated view of complex cell communication networks. In this review, I intend to provide a critical perspective about the main mechanisms contributing to regulate cellular crosstalk in the heart, which may be considered in the development of future therapeutic strategies, using cell-based therapies as a paradigmatic example. This Review has an associated Future Leader to Watch interview with the author.
Collapse
Affiliation(s)
- Tania Martins-Marques
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, 3000-548 Coimbra, Portugal
- Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Centre of Coimbra (CACC), 3004-561 Coimbra, Portugal
| |
Collapse
|
19
|
Janjusevic M, Fluca AL, Ferro F, Gagno G, D’Alessandra Y, Beltrami AP, Sinagra G, Aleksova A. Traditional and Emerging Biomarkers in Asymptomatic Left Ventricular Dysfunction-Promising Non-Coding RNAs and Exosomes as Biomarkers in Early Phases of Cardiac Damage. Int J Mol Sci 2021; 22:ijms22094937. [PMID: 34066533 PMCID: PMC8125492 DOI: 10.3390/ijms22094937] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022] Open
Abstract
Heart failure (HF) is one of the major causes of morbidity and mortality worldwide and represents an escalating problem for healthcare systems. The identification of asymptomatic patients with underlying cardiac subclinical disease would create an opportunity for early intervention and prevention of symptomatic HF. Traditional biomarkers are very useful as diagnostic and prognostic tools in the cardiovascular field; however, their application is usually limited to overt cardiac disease. On the other hand, a growing number of studies is investigating the diagnostic and prognostic potential of new biomarkers, such as micro-RNAs (miRNA), long non-coding RNAs, and exosome cargo, because of their involvement in the early phases of cardiac dysfunction. Unfortunately, their use in asymptomatic phases remains a distant goal. The aim of this review is to gather the current knowledge of old and novel biomarkers in the early diagnosis of cardiac dysfunction in asymptomatic individuals.
Collapse
Affiliation(s)
- Milijana Janjusevic
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (M.J.); (A.L.F.); (F.F.); (G.G.); (G.S.)
| | - Alessandra Lucia Fluca
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (M.J.); (A.L.F.); (F.F.); (G.G.); (G.S.)
| | - Federico Ferro
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (M.J.); (A.L.F.); (F.F.); (G.G.); (G.S.)
| | - Giulia Gagno
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (M.J.); (A.L.F.); (F.F.); (G.G.); (G.S.)
| | - Yuri D’Alessandra
- Cardiovascular Proteomics Unit, Centro Cardiologico Monzino-IRCCS, Via Parea 4, 20138 Milan, Italy;
| | | | - Gianfranco Sinagra
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (M.J.); (A.L.F.); (F.F.); (G.G.); (G.S.)
| | - Aneta Aleksova
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (M.J.); (A.L.F.); (F.F.); (G.G.); (G.S.)
- Correspondence: or ; Tel.: +39-3405507762; Fax: +39-040-3994878
| |
Collapse
|
20
|
Hall C, Gehmlich K, Denning C, Pavlovic D. Complex Relationship Between Cardiac Fibroblasts and Cardiomyocytes in Health and Disease. J Am Heart Assoc 2021; 10:e019338. [PMID: 33586463 PMCID: PMC8174279 DOI: 10.1161/jaha.120.019338] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiac fibroblasts are the primary cell type responsible for deposition of extracellular matrix in the heart, providing support to the contracting myocardium and contributing to a myriad of physiological signaling processes. Despite the importance of fibrosis in processes of wound healing, excessive fibroblast proliferation and activation can lead to pathological remodeling, driving heart failure and the onset of arrhythmias. Our understanding of the mechanisms driving the cardiac fibroblast activation and proliferation is expanding, and evidence for their direct and indirect effects on cardiac myocyte function is accumulating. In this review, we focus on the importance of the fibroblast-to-myofibroblast transition and the cross talk of cardiac fibroblasts with cardiac myocytes. We also consider the current use of models used to explore these questions.
Collapse
Affiliation(s)
- Caitlin Hall
- Institute of Cardiovascular Sciences University of Birmingham United Kingdom
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences University of Birmingham United Kingdom.,Division of Cardiovascular Medicine Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford University of Oxford United Kingdom
| | - Chris Denning
- Biodiscovery Institute University of Nottingham United Kingdom
| | - Davor Pavlovic
- Institute of Cardiovascular Sciences University of Birmingham United Kingdom
| |
Collapse
|
21
|
Nguyen BY, Azam T, Wang X. Cellular signaling cross-talk between different cardiac cell populations: an insight into the role of exosomes in the heart diseases and therapy. Am J Physiol Heart Circ Physiol 2021; 320:H1213-H1234. [PMID: 33513083 DOI: 10.1152/ajpheart.00718.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Exosomes are a subgroup of extracellular bilayer membrane nanovesicles that are enriched in a variety of bioactive lipids, receptors, transcription factors, surface proteins, DNA, and noncoding RNAs. They have been well recognized to play essential roles in mediating intercellular signaling by delivering bioactive molecules from host cells to regulate the physiological processes of recipient cells. In the context of heart diseases, accumulating studies have indicated that exosome-carried cellular proteins and noncoding RNA derived from different types of cardiac cells, including cardiomyocytes, fibroblasts, endothelial cells, immune cells, adipocytes, and resident stem cells, have pivotal roles in cardiac remodeling under disease conditions such as cardiac hypertrophy, diabetic cardiomyopathy, and myocardial infarction. In addition, exosomal contents derived from stem cells have been shown to be beneficial for regenerative potential of the heart. In this review, we discuss current understanding of the role of exosomes in cardiac communication, with a focus on cardiovascular pathophysiology and perspectives for their potential uses as cardiac therapies.
Collapse
Affiliation(s)
- Binh Yen Nguyen
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Tayyiba Azam
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Xin Wang
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
22
|
Xue R, Tan W, Wu Y, Dong B, Xie Z, Huang P, He J, Dong Y, Liu C. Role of Exosomal miRNAs in Heart Failure. Front Cardiovasc Med 2020; 7:592412. [PMID: 33392270 PMCID: PMC7773699 DOI: 10.3389/fcvm.2020.592412] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022] Open
Abstract
Heart failure is the terminal outcome of the majority of cardiovascular diseases, which lacks specific diagnostic biomarkers and therapeutic targets. It contributes to most of cardiovascular hospitalizations and death despite of the current therapy. Therefore, it is important to explore potential molecules improving the diagnosis and treatment of heart failure. MicroRNAs (miRNAs) are small non-coding RNAs that have been reported to be involved in regulating processes of heart failure. After the discovery of miRNAs in exosomes, the subcellular distribution analysis of miRNAs is raising researchers' attention. Growing evidence demonstrates that exosomal miRNAs may be promising diagnostic and therapeutic molecules for heart failure. This review summarizes the role of exosomal miRNAs in heart failure in the prospect of molecular and clinical researches.
Collapse
Affiliation(s)
- Ruicong Xue
- Department of Cardiology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Weiping Tan
- Department of Respiratory, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuzhong Wu
- Department of Cardiology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bin Dong
- Department of Cardiology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zengshuo Xie
- Department of Cardiology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Peisen Huang
- Department of Cardiology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiangui He
- Department of Cardiology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yugang Dong
- Department of Cardiology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chen Liu
- Department of Cardiology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
23
|
Lin Y, Anderson JD, Rahnama LMA, Gu SV, Knowlton AA. Exosomes in disease and regeneration: biological functions, diagnostics, and beneficial effects. Am J Physiol Heart Circ Physiol 2020; 319:H1162-H1180. [PMID: 32986962 PMCID: PMC7792703 DOI: 10.1152/ajpheart.00075.2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 07/30/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022]
Abstract
Exosomes are a subtype of extracellular vesicles. They range from 30 to 150 nm in diameter and originate from intraluminal vesicles. Exosomes were first identified as the mechanism for releasing unnecessary molecules from reticulocytes as they matured to red blood cells. Since then, exosomes have been shown to be secreted by a broad spectrum of cells and play an important role in the cardiovascular system. Different stimuli are associated with increased exosome release and result in different exosome content. The release of harmful DNA and other molecules via exosomes has been proposed as a mechanism to maintain cellular homeostasis. Because exosomes contain parent cell-specific proteins on the membrane and in the cargo that is delivered to recipient cells, exosomes are potential diagnostic biomarkers of various types of diseases, including cardiovascular disease. As exosomes are readily taken up by other cells, stem cell-derived exosomes have been recognized as a potential cell-free regenerative therapy to repair not only the injured heart but other tissues as well. The objective of this review is to provide an overview of the biological functions of exosomes in heart disease and tissue regeneration. Therefore, state-of-the-art methods for exosome isolation and characterization, as well as approaches to assess exosome functional properties, are reviewed. Investigation of exosomes provides a new approach to the study of disease and biological processes. Exosomes provide a potential "liquid biopsy," as they are present in most, if not all, biological fluids that are released by a wide range of cell types.
Collapse
Affiliation(s)
- Yun Lin
- Molecular and Cellular Cardiology, Cardiovascular Medicine, University of California, Davis, California
| | | | - Lily M A Rahnama
- Molecular and Cellular Cardiology, Cardiovascular Medicine, University of California, Davis, California
| | - Shenwen V Gu
- Molecular and Cellular Cardiology, Cardiovascular Medicine, University of California, Davis, California
| | - Anne A Knowlton
- Molecular and Cellular Cardiology, Cardiovascular Medicine, University of California, Davis, California
| |
Collapse
|
24
|
Yoshida T, Das NA, Carpenter AJ, Izadpanah R, Kumar SA, Gautam S, Bender SB, Siebenlist U, Chandrasekar B. Minocycline reverses IL-17A/TRAF3IP2-mediated p38 MAPK/NF-κB/iNOS/NO-dependent cardiomyocyte contractile depression and death. Cell Signal 2020; 73:109690. [PMID: 32553549 DOI: 10.1016/j.cellsig.2020.109690] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 12/11/2022]
Abstract
Minocycline, an FDA-approved second-generation semisynthetic tetracycline, exerts antioxidant, anti-apoptotic and anti-inflammatory effects, independent of its antimicrobial properties. Interleukin (IL)-17A is an immune and inflammatory mediator, and its sustained induction is associated with various cardiovascular diseases. Here we investigated (i) whether IL-17A induces cardiomyocyte contractile depression and death, (ii) whether minocycline reverses IL-17A's negative inotropic effects and (iii) investigated the underlying molecular mechanisms. Indeed, treatment with recombinant mouse IL-17A impaired adult cardiomyocyte contractility as evidenced by a 34% inhibition in maximal velocity of shortening and relengthening after 4 h (P < .01). Contractile depression followed iNOS induction at 2 h (2.13-fold, P < .01) and NO generation at 3 h (3.71-fold, P <.01). Further mechanistic investigations revealed that IL-17A-dependent induction of iNOS occurred via TRAF3IP2, TRAF6, TAK1, NF-κB, and p38MAPK signaling. 1400 W, a highly specific iNOS inhibitor, suppressed IL-17A-induced NO generation and contractile depression, where as the NO donors SNAP and PAPA-NONOate both suppressed cardiomyocyte contractility. IL-17A also stimulated cardiomyocyte IL-1β and TNF-α secretion, however, their neutralization failed to modulate IL-17A-mediated contractile depression or viability. Further increases of IL-17A concentration and the duration of exposure enhanced IL-1β and TNF-α secreted levels, buthad no impact on adult cardiomyocyte viability. However, when combined with pathophysiological concentrations of IL-1β or TNF-α, IL-17A promoted adult cardiomyocyte death. Importantly, minocycline blunted IL-17A-mediated deleterious effects, indicating its therapeutic potential in inflammatory cardiac diseases.
Collapse
Affiliation(s)
- Tadashi Yoshida
- Medicine/Heart and Vascular Institute, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Nitin A Das
- Cardiothoracic Surgery, UT Health, San Antonio, TX 78229, USA
| | | | - Reza Izadpanah
- Medicine/Heart and Vascular Institute, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Senthil A Kumar
- Medicine/Cardiovascular Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Sandeep Gautam
- Medicine/Cardiovascular Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Shawn B Bender
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO 65201, USA; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA; Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Ulrich Siebenlist
- Laboratory of Molecular Immunology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Bysani Chandrasekar
- Medicine/Cardiovascular Medicine, University of Missouri, Columbia, MO 65211, USA; Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO 65201, USA; Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65211, USA; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA.
| |
Collapse
|
25
|
Orozco P, Montoya Y, Bustamante J. Development of endomyocardial fibrosis model using a cell patterning technique: In vitro interaction of cell coculture of 3T3 fibroblasts and RL-14 cardiomyocytes. PLoS One 2020; 15:e0229158. [PMID: 32092082 PMCID: PMC7039516 DOI: 10.1371/journal.pone.0229158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/30/2020] [Indexed: 01/07/2023] Open
Abstract
Cardiac functions can be altered by changes in the microstructure of the heart, i.e., remodeling of the cardiac tissue, which may activate pathologies such as hypertrophy, dilation, or cardiac fibrosis. Cardiac fibrosis can develop due to an excessive deposition of extracellular matrix proteins, which are products of the activation of fibroblasts. In this context, the anatomical-histological change may interfere with the functioning of the cardiac tissue, which requires specialized cells for its operation. The purpose of the present study was to determine the cellular interactions and morphological changes in cocultures of 3T3 fibroblasts and RL-14 cardiomyocytes via the generation of a platform an in vitro model. For this purpose, a platform emulating the biological characteristics of endomyocardial fibrosis was generated using a cell patterning technique to study morphological cellular changes in compact and irregular patterns of fibrosis. It was found that cellular patterns emulating the geometrical distributions of endomyocardial fibrosis generated morphological changes after interaction of the RL-14 cardiomyocytes with the 3T3 fibroblasts. Through this study, it was possible to evaluate biological characteristics such as cell proliferation, adhesion, and spatial distribution, which are directly related to the type of emulated endomyocardial fibrosis. This research concluded that fibroblasts inhibited the proliferation of cardiomyocytes via their interaction with specific microarchitectures. This behavior is consistent with the histopathological distribution of cardiac fibrosis; therefore, the platform developed in this research could be useful for the in vitro assessment of cellular microdomains. This would allow for the experimental determination of interactions with drugs, substrates, or biomaterials within the engineering of cardiac tissues.
Collapse
Affiliation(s)
- Paola Orozco
- Centro de Bioingeniería, Grupo de Dinámica Cardiovascular, Universidad Pontificia Bolivariana, Medellín, Colombia
| | - Yuliet Montoya
- Centro de Bioingeniería, Grupo de Dinámica Cardiovascular, Universidad Pontificia Bolivariana, Medellín, Colombia
- Comité de Trabajo de Bioingeniería Cardiovascular, Sociedad Colombiana de Cardiología y Cirugía Cardiovascular, Bogotá, Colombia
| | - John Bustamante
- Centro de Bioingeniería, Grupo de Dinámica Cardiovascular, Universidad Pontificia Bolivariana, Medellín, Colombia
- Comité de Trabajo de Bioingeniería Cardiovascular, Sociedad Colombiana de Cardiología y Cirugía Cardiovascular, Bogotá, Colombia
| |
Collapse
|
26
|
Beauchamp P, Jackson CB, Ozhathil LC, Agarkova I, Galindo CL, Sawyer DB, Suter TM, Zuppinger C. 3D Co-culture of hiPSC-Derived Cardiomyocytes With Cardiac Fibroblasts Improves Tissue-Like Features of Cardiac Spheroids. Front Mol Biosci 2020; 7:14. [PMID: 32118040 PMCID: PMC7033479 DOI: 10.3389/fmolb.2020.00014] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/24/2020] [Indexed: 12/16/2022] Open
Abstract
Purpose: Both cardiomyocytes and cardiac fibroblasts (CF) play essential roles in cardiac development, function, and remodeling. Properties of 3D co-cultures are incompletely understood. Hence, 3D co-culture of cardiomyocytes and CF was characterized, and selected features compared with single-type and 2D culture conditions. Methods: Human cardiomyocytes derived from induced-pluripotent stem cells (hiPSC-CMs) were obtained from Cellular Dynamics or Ncardia, and primary human cardiac fibroblasts from ScienCell. Cardiac spheroids were investigated using cryosections and whole-mount confocal microscopy, video motion analysis, scanning-, and transmission-electron microscopy (SEM, TEM), action potential recording, and quantitative PCR (qPCR). Results: Spheroids formed in hanging drops or in non-adhesive wells showed spontaneous contractions for at least 1 month with frequent media changes. SEM of mechanically opened spheroids revealed a dense inner structure and no signs of blebbing. TEM of co-culture spheroids at 1 month showed myofibrils, intercalated disc-like structures and mitochondria. Ultrastructural features were comparable to fetal human myocardium. We then assessed immunostained 2D cultures, cryosections of spheroids, and whole-mount preparations by confocal microscopy. CF in co-culture spheroids assumed a small size and shape similar to the situation in ventricular tissue. Spheroids made only of CF and cultured for 3 weeks showed no stress fibers and strongly reduced amounts of alpha smooth muscle actin compared to early spheroids and 2D cultures as shown by confocal microscopy, western blotting, and qPCR. The addition of CF to cardiac spheroids did not lead to arrhythmogenic effects as measured by sharp-electrode electrophysiology. Video motion analysis showed a faster spontaneous contraction rate in co-culture spheroids compared to pure hiPSC-CMs, but similar contraction amplitudes and kinetics. Spontaneous contraction rates were not dependent on spheroid size. Applying increasing pacing frequencies resulted in decreasing contraction amplitudes without positive staircase effect. Gene expression analysis of selected cytoskeleton and myofibrillar proteins showed more tissue-like expression patterns in co-culture spheroids than with cardiomyocytes alone or in 2D culture. Conclusion: We demonstrate that the use of 3D co-culture of hiPSC-CMs and CF is superior over 2D culture conditions for co-culture models and more closely mimicking the native state of the myocardium with relevance to drug development as well as for personalized medicine.
Collapse
Affiliation(s)
- Philippe Beauchamp
- Cardiology Department, DBMR MEM C812, Bern University Hospital, Bern, Switzerland
| | - Christopher B. Jackson
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- IKELOS GmbH, Bern, Switzerland
| | | | | | - Cristi L. Galindo
- Division of Cardiovascular Medicine, Vanderbilt University Medical School, Nashville, TN, United States
- Department of Cell Biology and Molecular Biology, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Douglas B. Sawyer
- Department of Cardiac Services, Maine Medical Center, Scarborough, ME, United States
| | - Thomas M. Suter
- Cardiology Department, DBMR MEM C812, Bern University Hospital, Bern, Switzerland
| | - Christian Zuppinger
- Cardiology Department, DBMR MEM C812, Bern University Hospital, Bern, Switzerland
| |
Collapse
|
27
|
Abstract
Cardiac fibrosis is a pathological condition that occurs after injury and during aging. Currently, there are limited means to effectively reduce or reverse fibrosis. Key to identifying methods for curbing excess deposition of extracellular matrix is a better understanding of the cardiac fibroblast, the cell responsible for collagen production. In recent years, the diversity and functions of these enigmatic cells have been gradually revealed. In this review, I outline current approaches for identifying and classifying cardiac fibroblasts. An emphasis is placed on new insights into the heterogeneity of these cells as determined by lineage tracing and single-cell sequencing in development, adult, and disease states. These recent advances in our understanding of the fibroblast provide a platform for future development of novel therapeutics to combat cardiac fibrosis.
Collapse
Affiliation(s)
- Michelle D Tallquist
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii 96813, USA;
| |
Collapse
|
28
|
McArthur L, Riddell A, Chilton L, Smith GL, Nicklin SA. Regulation of connexin 43 by interleukin 1β in adult rat cardiac fibroblasts and effects in an adult rat cardiac myocyte: fibroblast co-culture model. Heliyon 2019; 6:e03031. [PMID: 31909243 PMCID: PMC6940628 DOI: 10.1016/j.heliyon.2019.e03031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/22/2019] [Accepted: 12/10/2019] [Indexed: 01/05/2023] Open
Abstract
Connexin 43 expression (Cx43) is increased in cardiac fibroblasts (CFs) following myocardial infarction. Here, potential mediators responsible for increasing Cx43 expression and effects of differential CF phenotype on cardiac myocyte (CM) function were investigated. Stimulating adult rat CFs with proinflammatory mediators revealed that interleukin 1β (IL-1β) significantly enhanced Cx43 levels through the IL-1β pathway. Additionally, IL-1β reduced mRNA levels of the myofibroblast (MF) markers: (i) connective tissue growth factor (CTGF) and (ii) α smooth muscle actin (αSMA), compared to control CFs. A co-culture adult rat CM:CF model was utilised to examine cell-to-cell interactions. Transfer of calcein from CMs to underlying CFs suggested functional gap junction formation. Functional analysis revealed contraction duration (CD) of CMs was shortened in co-culture with CFs, while treatment of CFs with IL-1β reduced this mechanical effect of co-culture. No effect on action potential rise time or duration of CMs cultured with control or IL-1β-treated CFs was observed. These data demonstrate that stimulating CFs with IL-1β increases Cx43 and reduces MF marker expression, suggesting altered cell phenotype. These changes may underlie the reduced mechanical effects of IL-1β treated CFs on CD of co-cultured CMs and therefore have an implication for our understanding of heterocellular interactions in cardiac disease.
Collapse
Affiliation(s)
- Lisa McArthur
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Alexandra Riddell
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Lisa Chilton
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Queensland, Australia
| | - Godfrey L Smith
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Stuart A Nicklin
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| |
Collapse
|
29
|
Patil M, Henderson J, Luong H, Annamalai D, Sreejit G, Krishnamurthy P. The Art of Intercellular Wireless Communications: Exosomes in Heart Disease and Therapy. Front Cell Dev Biol 2019; 7:315. [PMID: 31850349 PMCID: PMC6902075 DOI: 10.3389/fcell.2019.00315] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022] Open
Abstract
Exosomes are nanoscale membrane-bound extracellular vesicles secreted by most eukaryotic cells in the body that facilitates intercellular communication. Exosomes carry several signaling biomolecules, including miRNA, proteins, enzymes, cell surface receptors, growth factors, cytokines and lipids that can modulate target cell biology and function. Due to these capabilities, exosomes have emerged as novel intercellular signaling mediators in both homeostasis and pathophysiological conditions. Recent studies document that exosomes (both circulating or released from heart tissue) have been actively involved in cardiac remodeling in response to stressors. Also, exosomes released from progenitor/stem cells have protective effects in heart diseases and shown to have regenerative potential in the heart. In this review we discuss- the critical role played by circulating exosomes released from various tissues and from cells within the heart in cardiac health; the gap in knowledge that needs to be addressed to promote future research; and exploitation of recent advances in exosome engineering to develop novel therapy.
Collapse
Affiliation(s)
- Mallikarjun Patil
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - John Henderson
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Hien Luong
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Divya Annamalai
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Gopalkrishna Sreejit
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
30
|
Blythe NM, Muraki K, Ludlow MJ, Stylianidis V, Gilbert HTJ, Evans EL, Cuthbertson K, Foster R, Swift J, Li J, Drinkhill MJ, van Nieuwenhoven FA, Porter KE, Beech DJ, Turner NA. Mechanically activated Piezo1 channels of cardiac fibroblasts stimulate p38 mitogen-activated protein kinase activity and interleukin-6 secretion. J Biol Chem 2019; 294:17395-17408. [PMID: 31586031 PMCID: PMC6873183 DOI: 10.1074/jbc.ra119.009167] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 10/02/2019] [Indexed: 12/03/2022] Open
Abstract
Piezo1 is a mechanosensitive cation channel with widespread physiological importance; however, its role in the heart is poorly understood. Cardiac fibroblasts help preserve myocardial integrity and play a key role in regulating its repair and remodeling following stress or injury. Here we investigated Piezo1 expression and function in cultured human and mouse cardiac fibroblasts. RT-PCR experiments confirmed that Piezo1 mRNA in cardiac fibroblasts is expressed at levels similar to those in endothelial cells. The results of a Fura-2 intracellular Ca2+ assay validated Piezo1 as a functional ion channel that is activated by its agonist, Yoda1. Yoda1-induced Ca2+ entry was inhibited by Piezo1 blockers (gadolinium and ruthenium red) and was reduced proportionally by siRNA-mediated Piezo1 knockdown or in murine Piezo1+/- cells. Results from cell-attached patch clamp recordings on human cardiac fibroblasts established that they contain mechanically activated ion channels and that their pressure responses are reduced by Piezo1 knockdown. Investigation of Yoda1 effects on selected remodeling genes indicated that Piezo1 activation increases both mRNA levels and protein secretion of IL-6, a pro-hypertrophic and profibrotic cytokine, in a Piezo1-dependent manner. Moreover, Piezo1 knockdown reduced basal IL-6 expression from cells cultured on softer collagen-coated substrates. Multiplex kinase activity profiling combined with kinase inhibitor experiments and phosphospecific immunoblotting established that Piezo1 activation stimulates IL-6 secretion via the p38 mitogen-activated protein kinase downstream of Ca2+ entry. In summary, cardiac fibroblasts express mechanically activated Piezo1 channels coupled to secretion of the paracrine signaling molecule IL-6. Piezo1 may therefore be important in regulating cardiac remodeling.
Collapse
Affiliation(s)
- Nicola M Blythe
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Katsuhiko Muraki
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, 1-100 Kusumoto, Chikusa, Nagoya 464-8650, Japan
| | - Melanie J Ludlow
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Vasili Stylianidis
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, United Kingdom
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht 6200MD, The Netherlands
| | - Hamish T J Gilbert
- Wellcome Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of[c27c]áBiological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester,[c27c]áM13 9PL, United Kingdom
| | - Elizabeth L Evans
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Kevin Cuthbertson
- School of Chemistry, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Richard Foster
- School of Chemistry, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Joe Swift
- Wellcome Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of[c27c]áBiological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester,[c27c]áM13 9PL, United Kingdom
| | - Jing Li
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Mark J Drinkhill
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Frans A van Nieuwenhoven
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht 6200MD, The Netherlands
| | - Karen E Porter
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - David J Beech
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Neil A Turner
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
31
|
Appiah C, Arndt C, Siemsen K, Heitmann A, Staubitz A, Selhuber-Unkel C. Living Materials Herald a New Era in Soft Robotics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1807747. [PMID: 31267628 DOI: 10.1002/adma.201807747] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/07/2019] [Indexed: 05/22/2023]
Abstract
Living beings have an unsurpassed range of ways to manipulate objects and interact with them. They can make autonomous decisions and can heal themselves. So far, a conventional robot cannot mimic this complexity even remotely. Classical robots are often used to help with lifting and gripping and thus to alleviate the effects of menial tasks. Sensors can render robots responsive, and artificial intelligence aims at enabling autonomous responses. Inanimate soft robots are a step in this direction, but it will only be in combination with living systems that full complexity will be achievable. The field of biohybrid soft robotics provides entirely new concepts to address current challenges, for example the ability to self-heal, enable a soft touch, or to show situational versatility. Therefore, "living materials" are at the heart of this review. Similarly to biological taxonomy, there is a recent effort for taxonomy of biohybrid soft robotics. Here, an expansion is proposed to take into account not only function and origin of biohybrid soft robotic components, but also the materials. This materials taxonomy key demonstrates visually that materials science will drive the development of the field of soft biohybrid robotics.
Collapse
Affiliation(s)
- Clement Appiah
- Institute for Organic and Analytical Chemistry, University of Bremen, Leobener Str. 7, D-28359, Bremen, Germany
- MAPEX Center for Materials and Processes, University of Bremen, Bibliothekstraße 1, D-28359, Bremen, Germany
| | - Christine Arndt
- Institute for Materials Science, University of Kiel, Kaiserstr. 2, D-24143, Kiel, Germany
| | - Katharina Siemsen
- Institute for Materials Science, University of Kiel, Kaiserstr. 2, D-24143, Kiel, Germany
| | - Anne Heitmann
- Institute for Organic and Analytical Chemistry, University of Bremen, Leobener Str. 7, D-28359, Bremen, Germany
- MAPEX Center for Materials and Processes, University of Bremen, Bibliothekstraße 1, D-28359, Bremen, Germany
| | - Anne Staubitz
- Institute for Organic and Analytical Chemistry, University of Bremen, Leobener Str. 7, D-28359, Bremen, Germany
- MAPEX Center for Materials and Processes, University of Bremen, Bibliothekstraße 1, D-28359, Bremen, Germany
- Otto-Diels-Institute for Organic Chemistry, University of Kiel, Otto-Hahn-Platz 4, D-24118, Kiel, Germany
| | | |
Collapse
|
32
|
Wu Y, Qin YH, Liu Y, Zhu L, Zhao XX, Liu YY, Luo SW, Tang GS, Shen Q. Cardiac troponin I autoantibody induces myocardial dysfunction by PTEN signaling activation. EBioMedicine 2019; 47:329-340. [PMID: 31474552 PMCID: PMC6796505 DOI: 10.1016/j.ebiom.2019.08.045] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/02/2019] [Accepted: 08/21/2019] [Indexed: 01/22/2023] Open
Abstract
Background The objective of the current study was to study the molecular mechanism(s) underlying cardiac troponin I autoantibody (cTnIAAb) binding to cardiomyocyte and resultant myocardial damage/dysfunction. Methods cTnIAAb was purified from serum of 10 acute myocardial infarction (AMI) patients with left ventricular remodeling. Recombinant human cTnI was used to generate three mouse-derived monoclonal anti-cTnI antibodies (cTnImAb1, cTnImAb2, and cTnImAb3). The target proteins in cardiac myocyte membrane bound to cTnImAb and effect of cTnIAAb and cTnImAb on apoptosis and myocardial function were determined. Findings We found that cTnIAAb/cTnImAb1 directly bound to the cardiomyocyte membraneα-Enolase (ENO1) and triggered cell apoptosis via increased expression of ENO1 and Bax, decreased expression of Bcl2, subsequently activating Caspase8, Caspase 3, phosphatase and tensin homolog (PTEN) while inhibiting Akt activity. This cTnIAAb-ENO1-PTEN-Akt signaling axis contributed to increased myocardial apoptosis, myocardial collagen deposition, and impaired systolic dysfunction. Interpretation Results obtained in this study indicate that cTnIAAb is involved in the process of ventricular remodeling after myocardial injury. Fund The National Natural Science Foundation of China (Grant#: 81260026).
Collapse
Affiliation(s)
- Yu Wu
- Outpatient Department, Changcheng Hospital, Nanchang University, Nanchang, Jiangxi 330002, China; Department of Laboratory Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yang-Hua Qin
- Department of Laboratory Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yang Liu
- Department of Cardiothoracic Surgery, Changhai Hospital, Second military Medical University, Shanghai 200433, China
| | - Li Zhu
- Department of Laboratory Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, China; Department of Laboratory Medicine, Wuxi First People Hospital, Wuxi, Jiangsu 214002, China
| | - Xian-Xian Zhao
- Department of Cardiology, Changhai Hospital, Second military Medical University, Shanghai 200433, China
| | - Yao-Yang Liu
- Department of Rheumatology, Changzheng Hospital, Second military Medical University, Shanghai 200003, China
| | - Shi-Wen Luo
- Research Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Gu-Sheng Tang
- Department of Hematology, Changhai Hospital, Second military Medical University, Shanghai 200433, China.
| | - Qian Shen
- Department of Laboratory Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
33
|
Leitolis A, Robert AW, Pereira IT, Correa A, Stimamiglio MA. Cardiomyogenesis Modeling Using Pluripotent Stem Cells: The Role of Microenvironmental Signaling. Front Cell Dev Biol 2019; 7:164. [PMID: 31448277 PMCID: PMC6695570 DOI: 10.3389/fcell.2019.00164] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 07/29/2019] [Indexed: 12/20/2022] Open
Abstract
Pluripotent stem cells (PSC) can be used as a model to study cardiomyogenic differentiation. In vitro modeling can reproduce cardiac development through modulation of some key signaling pathways. Therefore, many studies make use of this strategy to better understand cardiomyogenesis complexity and to determine possible ways to modulate cell fate. However, challenges remain regarding efficiency of differentiation protocols, cardiomyocyte (CM) maturation and therapeutic applications. Considering that the extracellular milieu is crucial for cellular behavior control, cardiac niche studies, such as those identifying secreted molecules from adult or neonatal tissues, allow the identification of extracellular factors that may contribute to CM differentiation and maturation. This review will focus on cardiomyogenesis modeling using PSC and the elements involved in cardiac microenvironmental signaling (the secretome - extracellular vesicles, extracellular matrix and soluble factors) that may contribute to CM specification and maturation.
Collapse
Affiliation(s)
- Amanda Leitolis
- Stem Cell Basic Biology Laboratory, Carlos Chagas Institute, FIOCRUZ-PR, Curitiba, Brazil
| | - Anny W Robert
- Stem Cell Basic Biology Laboratory, Carlos Chagas Institute, FIOCRUZ-PR, Curitiba, Brazil
| | - Isabela T Pereira
- Stem Cell Basic Biology Laboratory, Carlos Chagas Institute, FIOCRUZ-PR, Curitiba, Brazil
| | - Alejandro Correa
- Stem Cell Basic Biology Laboratory, Carlos Chagas Institute, FIOCRUZ-PR, Curitiba, Brazil
| | - Marco A Stimamiglio
- Stem Cell Basic Biology Laboratory, Carlos Chagas Institute, FIOCRUZ-PR, Curitiba, Brazil
| |
Collapse
|
34
|
Nakanishi H, Lee JK, Miwa K, Masuyama K, Yasutake H, Li J, Tomoyama S, Honda Y, Deguchi J, Tsujimoto S, Hidaka K, Miyagawa S, Sawa Y, Komuro I, Sakata Y. Geometrical Patterning and Constituent Cell Heterogeneity Facilitate Electrical Conduction Disturbances in a Human Induced Pluripotent Stem Cell-Based Platform: An In vitro Disease Model of Atrial Arrhythmias. Front Physiol 2019; 10:818. [PMID: 31316396 PMCID: PMC6610482 DOI: 10.3389/fphys.2019.00818] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 06/11/2019] [Indexed: 01/09/2023] Open
Abstract
Ectopic foci from pulmonary veins (PVs) comprise the main trigger associated with the initiation of atrial fibrillation (AF). An abrupt anatomical narrow-to-wide transition, modeled as in vitro geometrical patterning with similar configuration in the present study, is located at the junction of PVs and the left atrium (LA). Complex cellular composition, i.e., constituent cell heterogeneity, is also observed in PVs and the PVs-LA junction. High frequency triggers accompanied with anatomical irregularity and constituent cell heterogeneity provoke impaired conduction, a prerequisite for AF genesis. However, few experiments investigating the effects of these factors on electrophysiological properties using human-based cardiomyocytes (CMs) with atrial properties have been reported. The aim of the current study was to estimate whether geometrical patterning and constituent cell heterogeneity under high frequency stimuli undergo conduction disturbance utilizing an in vitro two-dimensional (2D) monolayer preparation consisting of atrial-like CMs derived from human induced pluripotent stem cells (hiPSCs) and atrial fibroblasts (Fbs). We induced hiPSCs into atrial-like CMs using a directed cardiac differentiation protocol with the addition of all-trans retinoic acid (ATRA). The atrial-like hiPSC-derived CMs (hiPSC-CMs) and atrial Fbs were transferred in defined ratios (CMs/Fbs: 100%/0% or 70%/30%) on manually fabricated plates with or without geometrical patterning imitating the PVs-LA junction. High frequency field stimulation emulating repetitive ectopic foci originated in PVs were delivered, and the electrical propagation was assessed by optical mapping. We generated high purity CMs with or without the ATRA application. ATRA-treated hiPSC-CMs exhibited significantly higher atrial-specific properties by immunofluorescence staining, gene expression patterns, and optical action potential parameters than those of ATRA-untreated hiPSC-CMs. Electrical stimuli at a higher frequency preferentially induced impaired electrical conduction on atrial-like hiPSC-CMs monolayer preparations with an abrupt geometrical transition than on those with uniform geometry. Additionally, the application of human atrial Fbs to the geometrically patterned atrial-like hiPSC-CMs tended to further deteriorate the integrity of electrical conduction compared with those using the atrial-like hiPSC-CM alone preparations. Thus, geometrical narrow-to-wide patterning under high frequency stimuli preferentially jeopardized electrical conduction within in vitro atrial-like hiPSC-CM monolayers. Constituent cell heterogeneity represented by atrial Fbs also contributed to the further deterioration of conduction stability.
Collapse
Affiliation(s)
- Hiroyuki Nakanishi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Jong-Kook Lee
- Department of Advanced Cardiovascular Regenerative Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Keiko Miwa
- Department of Mechanical Engineering, Kyushu University, Fukuoka, Japan
| | - Kiyoshi Masuyama
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hideki Yasutake
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Jun Li
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Satoki Tomoyama
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yayoi Honda
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Jiro Deguchi
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Shinji Tsujimoto
- Regenerative & Cellular Medicine Office, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Kyoko Hidaka
- Department of Advanced Cardiovascular Regenerative Medicine, Graduate School of Medicine, Osaka University, Suita, Japan.,Center for Fundamental Education, The University of Kitakyushu, Kitakyushu, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
35
|
Beringer A, Miossec P. Systemic effects of IL-17 in inflammatory arthritis. Nat Rev Rheumatol 2019; 15:491-501. [PMID: 31227819 DOI: 10.1038/s41584-019-0243-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2019] [Indexed: 12/20/2022]
Abstract
Inflammatory arthritis occurs in many diseases and is characterized by joint inflammation and damage. However, the inflammatory state in arthritis is commonly associated with systemic manifestations, which are generally linked to a poor prognosis. The pro-inflammatory cytokine IL-17 functions within a complex network of cytokines and contributes to the pathogenesis of various inflammatory diseases. Three IL-17 inhibitors have already been approved for the treatment of psoriasis, psoriatic arthritis, and ankylosing spondylitis. After a brief description of IL-17 and its local effects on joints, this Review focuses on the systemic effects of IL-17 in inflammatory arthritis. Increased circulating concentrations of bioactive IL-17 mediate changes in blood vessels, liver and cardiac and skeletal muscles. The effects of IL-17 on vascular and cardiac cells might contribute to the increased risk of cardiovascular events that occurs in all patients with inflammatory disorders. In the liver, IL-17 contributes to the high circulating concentrations of acute-phase proteins, such as C-reactive protein, and the appearance of liver lesions. In skeletal muscle, IL-17 contributes to muscle contractibility defects and weakness. Thus, targeting IL-17 might have beneficial effects at both local and systemic levels, and could also be proposed for the treatment of a wider range of inflammatory diseases.
Collapse
Affiliation(s)
- Audrey Beringer
- Immunogenomics and Inflammation Research Unit, EA 4130, University of Lyon, Hospices Civils de Lyon, Lyon, France
| | - Pierre Miossec
- Immunogenomics and Inflammation Research Unit, EA 4130, University of Lyon, Hospices Civils de Lyon, Lyon, France.
| |
Collapse
|
36
|
Loonat AA, Curtis MK, Richards MA, Nunez-Alonso G, Michl J, Swietach P. A high-throughput ratiometric method for imaging hypertrophic growth in cultured primary cardiac myocytes. J Mol Cell Cardiol 2019; 130:184-196. [PMID: 30986378 PMCID: PMC6520438 DOI: 10.1016/j.yjmcc.2019.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/14/2019] [Accepted: 04/04/2019] [Indexed: 12/21/2022]
Abstract
Maladaptive hypertrophy of cardiac myocytes increases the risk of heart failure. The underlying signaling can be triggered and interrogated in cultured neonatal ventricular myocytes (NRVMs) using sophisticated pharmacological and genetic techniques. However, the methods for quantifying cell growth are, by comparison, inadequate. The lack of quantitative, calibratable and computationally-inexpensive high-throughput technology has limited the scope for using cultured myocytes in large-scale analyses. We present a ratiometric method for quantifying the hypertrophic growth of cultured myocytes, compatible with high-throughput imaging platforms. Protein biomass was assayed from sulforhodamine B (SRB) fluorescence, and image analysis calculated the quotient of signal from extra-nuclear and nuclear regions. The former readout relates to hypertrophic growth, whereas the latter is a reference for correcting protein-independent (e.g. equipment-related) variables. This ratiometric measure, when normalized to the number of cells, provides a robust quantification of cellular hypertrophy. The method was tested by comparing the efficacy of various chemical agonists to evoke hypertrophy, and verified using independent assays (myocyte area, transcripts of markers). The method's high resolving power and wide dynamic range were confirmed by the ability to generate concentration-response curves, track the time-course of hypertrophic responses with fine temporal resolution, describe drug/agonist interactions, and screen for novel anti-hypertrophic agents. The method can be implemented as an end-point in protocols investigating hypertrophy, and is compatible with automated plate-reader platforms for generating high-throughput data, thereby reducing investigator-bias. Finally, the computationally-minimal workflow required for obtaining measurements makes the method simple to implement in most laboratories. Maladaptive hypertrophy of myocytes can lead to heart failure. Common methods for tracking growth in cultured myocytes are inadequate. We design and test a method for tracking myocyte hypertrophy in vitro. The method provides a ratiometric index of growth for high throughput analyses. Using the method, we characterize further details of (anti)hypertrophic responses.
Collapse
Affiliation(s)
- Aminah A Loonat
- University of Oxford, Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, United Kingdom
| | - M Kate Curtis
- University of Oxford, Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Mark A Richards
- University of Oxford, Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Graciela Nunez-Alonso
- University of Oxford, Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Johanna Michl
- University of Oxford, Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Pawel Swietach
- University of Oxford, Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, United Kingdom.
| |
Collapse
|
37
|
Kaur K, Jalife J. Is TGF-β 1 (Transforming Growth Factor-β 1) an Enabler of Myofibroblast-Cardiomyocyte Cross Talk? Circ Arrhythm Electrophysiol 2019; 10:e005289. [PMID: 28500179 DOI: 10.1161/circep.117.005289] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Kuljeet Kaur
- From the Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor (K.K., J.J.); Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (J.J.); and Centro de Investigación Biomédica en Red, Enfermedades Cardiovasculares, Spain (J.J.)
| | - José Jalife
- From the Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor (K.K., J.J.); Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (J.J.); and Centro de Investigación Biomédica en Red, Enfermedades Cardiovasculares, Spain (J.J.).
| |
Collapse
|
38
|
Attenuation of doxorubicin-induced cardiotoxicity in a human in vitro cardiac model by the induction of the NRF-2 pathway. Biomed Pharmacother 2019; 112:108637. [PMID: 30798127 DOI: 10.1016/j.biopha.2019.108637] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 01/28/2019] [Accepted: 01/28/2019] [Indexed: 01/06/2023] Open
Abstract
Dose-dependent cardiotoxicity is the leading adverse reaction seen in cancer patients treated with doxorubicin. Currently, dexrazoxane is the only approved drug that can partially protect against this toxicity in patients, however, its administration is restricted to those patients receiving a high cumulative dose of anthracyclines. Investigations into the mechanisms of cardiotoxicity and efforts to improve cardioprotective strategies have been hindered by the limited availability of a phenotypically relevant in vitro adult human cardiac model system. Here, we adapted a readily reproducible, functional 3D human multi-cell type cardiac system to emulate patient responses seen with doxorubicin and dexrazoxane. We show that administration of two NRF2 gene inducers namely the semi-synthetic triterpenoid Bardoxolone methyl, and the isothiocyanate sulfurophane, result in cardioprotection against doxorubicin toxicity comparable to dexrazoxane as evidenced by an increase in cell viability and a decrease in the production of reactive oxygen species. We further show a synergistic attenuation of cardiotoxicity when the NRF2 inducers and dexrazoxane are used in tandem. Taken together, our data indicate that the 3D spheroid is a suitable model to investigate drug induced cardiotoxicity and we reveal an essential role of the NRF2 pathway in cardioprotection providing a novel pharmacological mechanism and intervention route towards the alleviation of doxorubicin-induced toxicity.
Collapse
|
39
|
Chu PY, Joshi MS, Horlock D, Kiriazis H, Kaye DM. CXCR4 Antagonism Reduces Cardiac Fibrosis and Improves Cardiac Performance in Dilated Cardiomyopathy. Front Pharmacol 2019; 10:117. [PMID: 30837882 PMCID: PMC6389782 DOI: 10.3389/fphar.2019.00117] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/31/2019] [Indexed: 01/05/2023] Open
Abstract
Background: Myocardial fibrosis is a key pathologic finding in the failing heart and is implicated as a cause of increased ventricular stiffness and susceptibility to ventricular arrhythmia. Neurohormonal mediators such as aldosterone and angiotensin II are known to cause fibrosis in experimental models, however, clinical evidence for the reversal of fibrosis with relevant antagonists is limited. Recent studies suggest that inflammatory mediators may contribute to fibrosis. In dilated cardiomyopathy the mechanism for myocardial fibrosis is unclear and its implications on systolic function are not known. Methods and Results: We studied the effect of a highly selective antagonist of SDF-1/CXCR4 signaling, AMD3100, on the development of cardiac fibrosis and cardiac function in mice with dilated cardiomyopathy due to cardiac-specific transgenic overexpression of the stress-kinase, Mst1. AMD3100 significantly attenuated the progression of myocardial fibrosis and this was accompanied by significant improvements in diastolic and systolic performance as evaluated in isolated Langendorff perfused hearts. AMD3100 reduced BNP mRNA expression but did not alter the expression of Ca2+ handling genes. CXCR4 antagonism also reduced the abundance of splenic CD4+ T cells. Conclusion: This study demonstrates that CXCR4 pathway contributes to pathogenesis of cardiac fibrosis in dilated cardiomyopathy, and it represents a new potential therapeutic target in heart failure. The data also demonstrate that anti-fibrotic strategies can improve systolic performance.
Collapse
Affiliation(s)
- Po-Yin Chu
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mandar S Joshi
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Duncan Horlock
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Helen Kiriazis
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - David M Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Medicine, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
40
|
Ma S, Ma J, Guo L, Bai J, Mao S, Zhang M. Tongguan capsule-derived herb reduces susceptibility to atrial fibrillation by inhibiting left atrial fibrosis via modulating cardiac fibroblasts. J Cell Mol Med 2019; 23:1197-1210. [PMID: 30456908 PMCID: PMC6349173 DOI: 10.1111/jcmm.14022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/11/2018] [Accepted: 10/16/2018] [Indexed: 12/19/2022] Open
Abstract
Tongguan capsule is a compound Chinese medicine used to treat ischaemic heart diseases. This study aimed to investigate whether Tongguan capsule-derived herb (TGD) has a preventive effect on atrial fibrillation (AF) in post-myocardial infarction (MI) rats and to determine the underlying mechanisms. MI was induced by ligation of the left anterior descending coronary artery. TGD was administered to the post-MI rats over a 4-week period. The TGD-treated rats had lower rates of AF inducibility and shorter AF durations than the MI rats. TGD improved the left atrial (LA) conduction velocity and homogeneity. It reduced the fibrosis-positive areas and the protein levels of collagen types I and III in the left atrium. In vitro, it inhibited the expression of collagen types I and III by inhibiting the proliferation, migration, differentiation and cytokine secretion of cardiac fibroblasts (CFs). In conclusion, the current study demonstrated that TGD reduces susceptibility to AF and improves LA conduction function in rats with post-MI by inhibiting left atrial fibrosis and modulating CFs. Targeting the CF population may be a novel antiarrhythmic therapeutic approach.
Collapse
Affiliation(s)
- Shiyu Ma
- Department of Critical‐care MedicineGuangdong Provincial Hospital of Chinese MedicineThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
- Guangzhou Key Laboratory of Myocardial Infarction in Chinese Medical Prevention and TreatmentGuangzhouChina
| | - Jin Ma
- Cardiac Electrophysiology Research TeamGuangdong Provincial Hospital of Chinese MedicineThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Liheng Guo
- Department of Critical‐care MedicineGuangdong Provincial Hospital of Chinese MedicineThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
- Guangzhou Key Laboratory of Myocardial Infarction in Chinese Medical Prevention and TreatmentGuangzhouChina
| | - Junqi Bai
- New Patent Chinese Medicine and Decoction Pieces Innovative Research and Development TeamThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangdong Provincial Hospital of Chinese MedicineGuangzhouChina
| | - Shuai Mao
- Department of Critical‐care MedicineGuangdong Provincial Hospital of Chinese MedicineThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
- Guangzhou Key Laboratory of Myocardial Infarction in Chinese Medical Prevention and TreatmentGuangzhouChina
| | - Minzhou Zhang
- Department of Critical‐care MedicineGuangdong Provincial Hospital of Chinese MedicineThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
- Guangzhou Key Laboratory of Myocardial Infarction in Chinese Medical Prevention and TreatmentGuangzhouChina
| |
Collapse
|
41
|
Zhang X, Zhu JX, Ma ZG, Wu HM, Xu SC, Song P, Kong CY, Yuan YP, Deng W, Tang QZ. Rosmarinic acid alleviates cardiomyocyte apoptosis via cardiac fibroblast in doxorubicin-induced cardiotoxicity. Int J Biol Sci 2019; 15:556-567. [PMID: 30745842 PMCID: PMC6367577 DOI: 10.7150/ijbs.29907] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 11/09/2018] [Indexed: 12/14/2022] Open
Abstract
Cardiomyocyte apoptosis is a key event in the process of doxorubicin (DOX)-induced cardiotoxicity. Our previous study found that rosmarinic acid (RA) could attenuate pressure overload-induced cardiac dysfunction via cardiac fibroblasts (CFs), however its effect in DOX-induced cardiotoxicity remains unknown. In the present study, mice were subjected to a single intraperitoneal injection of DOX (15mg/kg) to generate DOX-induced cardiotoxicity. Histological examination, echocardiography, and molecular markers were used to evaluate the effects of RA. Neonatal rat cardiomyocytes (CMs) and CFs were used to verify the protective effect of RA in vitro. Conditioned medium derived from RA-treated CFs were prepared to illustrate the effect of RA on paracrine interplay between CFs and CMs. We found that RA significantly alleviated DOX-induced cardiomyocyte apoptosis and cardiac dysfunction in vivo, which, however, had almost negligible beneficial effect on DOX directly induced cardiomyocyte apoptosis in vitro. Mechanistically, CFs-derived Fas L was responsible for DOX-induced cardiomyocyte apoptosis, and RA treatment could decrease Fas L expression in CFs and its release to the conditioned medium by suppressing nuclear factor of activated T cells (NFAT) activation and metalloproteinase 7 (MMP7) expression, and exerted the anti-apoptotic effect on CMs via CFs. Ionomycin, and activator of NFAT, abrogated RA-mediated protective effect on cardiomyocyte apoptosis and cardiac dysfunction. In summary, RA alleviated cardiomyocyte apoptosis by inhibiting the expression and release of Fas L in CFs via a paracrine manner, moreover, NFAT as well as MMP7 inhibition were responsible for the suppression of Fas L. RA could be a powerful new therapeutic agent to mitigate cardiomyocyte apoptosis, thereby improving DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Wei Deng
- ✉ Corresponding authors: Qi-Zhu Tang, Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, Wuhan University at Jiefang Road 238, Wuhan 430060, RP China. Tel.: +86 27 88073385; Fax: +86 27 88042292. E-mail: or Wei Deng, Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, Wuhan University at Jiefang Road 238, Wuhan 430060, RP China. Tel.: +86 27 88073385; Fax: +86 27 88042292. E-mail:
| | - Qi-Zhu Tang
- ✉ Corresponding authors: Qi-Zhu Tang, Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, Wuhan University at Jiefang Road 238, Wuhan 430060, RP China. Tel.: +86 27 88073385; Fax: +86 27 88042292. E-mail: or Wei Deng, Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, Wuhan University at Jiefang Road 238, Wuhan 430060, RP China. Tel.: +86 27 88073385; Fax: +86 27 88042292. E-mail:
| |
Collapse
|
42
|
Bollini S, Smits AM, Balbi C, Lazzarini E, Ameri P. Triggering Endogenous Cardiac Repair and Regeneration via Extracellular Vesicle-Mediated Communication. Front Physiol 2018; 9:1497. [PMID: 30405446 PMCID: PMC6206049 DOI: 10.3389/fphys.2018.01497] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 10/03/2018] [Indexed: 12/15/2022] Open
Abstract
A variety of paracrine signals create networks within the myocardium and mediate intercellular communications. Indeed, paracrine stimulation of the endogenous regenerative program of the heart, mainly based on resident cardiac progenitor cell (CPC) activation together with cardiomyocyte proliferation, has become increasingly relevant for future cardiac medicine. In the last years, it has been shown that extracellular vesicles (EV), including exosomes (Ex), are powerful conveyors of relevant biological effects. EV have been proposed not only as promising therapeutic tool for triggering cardiac regeneration and improving repair, but also as means of better understanding the physiological and pathological relationships between specific cardiac components, including cardiomyocytes and fibroblasts. Actually, EV from different kinds of exogenous stem cells have been shown to mediate beneficial effects on the injured myocardium. Moreover, endogenous cells, like CPC can instruct cardiovascular cell types, including cardiomyocytes, while cardiac stromal cells, especially fibroblasts, secrete EV that modulate relevant aspects of cardiomyocyte biology, such as hypertrophy and electrophysiological properties. Finally, cardiomyocytes too may release EV influencing the function of other cardiac cell types. Therefore, EV-based crosstalk is thought to be important in both physiology and pathology, being involved in the responses of the heart to noxious stimuli. In this review we will discuss the role of EV in both regulating cardiac homeostasis and driving heart regeneration. In particular, we will address their role in: (i) providing cardio-protection and enhancing cardiac repair mechanisms; (ii) CPC biology; and (iii) influencing adult cardiomyocyte behavior.
Collapse
Affiliation(s)
- Sveva Bollini
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genoa, Italy
| | - Anke M Smits
- Laboratory of Cardiovascular Cell Biology, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Carolina Balbi
- Laboratory of Molecular and Cellular Cardiology, CardioCentro Ticino, Lugano, Switzerland
| | - Edoardo Lazzarini
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genoa, Italy
| | - Pietro Ameri
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genoa, Italy.,Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
43
|
Antes TJ, Middleton RC, Luther KM, Ijichi T, Peck KA, Liu WJ, Valle J, Echavez AK, Marbán E. Targeting extracellular vesicles to injured tissue using membrane cloaking and surface display. J Nanobiotechnology 2018; 16:61. [PMID: 30165851 PMCID: PMC6116387 DOI: 10.1186/s12951-018-0388-4] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 08/16/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) and exosomes are nano-sized, membrane-bound vesicles shed by most eukaryotic cells studied to date. EVs play key signaling roles in cellular development, cancer metastasis, immune modulation and tissue regeneration. Attempts to modify exosomes to increase their targeting efficiency to specific tissue types are still in their infancy. Here we describe an EV membrane anchoring platform termed "cloaking" to directly embed tissue-specific antibodies or homing peptides on EV membrane surfaces ex vivo for enhanced vesicle uptake in cells of interest. The cloaking system consists of three components: DMPE phospholipid membrane anchor, polyethylene glycol spacer and a conjugated streptavidin platform molecule, to which any biotinylated molecule can be coupled for EV decoration. RESULTS We demonstrate the utility of membrane surface engineering and biodistribution tracking with this technology along with targeting EVs for enhanced uptake in cardiac fibroblasts, myoblasts and ischemic myocardium using combinations of fluorescent tags, tissue-targeting antibodies and homing peptide surface cloaks. We compare cloaking to a complementary approach, surface display, in which parental cells are engineered to secrete EVs with fusion surface targeting proteins. CONCLUSIONS EV targeting can be enhanced both by cloaking and by surface display; the former entails chemical modification of preformed EVs, while the latter requires genetic modification of the parent cells. Reduction to practice of the cloaking approach, using several different EV surface modifications to target distinct cells and tissues, supports the notion of cloaking as a platform technology.
Collapse
Affiliation(s)
- Travis J. Antes
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis Building, Los Angeles, CA 90048 USA
| | - Ryan C. Middleton
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis Building, Los Angeles, CA 90048 USA
| | - Kristin M. Luther
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis Building, Los Angeles, CA 90048 USA
| | - Takeshi Ijichi
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis Building, Los Angeles, CA 90048 USA
| | - Kiel A. Peck
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis Building, Los Angeles, CA 90048 USA
| | - Weixin Jane Liu
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis Building, Los Angeles, CA 90048 USA
| | - Jackie Valle
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis Building, Los Angeles, CA 90048 USA
| | - Antonio K. Echavez
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis Building, Los Angeles, CA 90048 USA
| | - Eduardo Marbán
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis Building, Los Angeles, CA 90048 USA
| |
Collapse
|
44
|
Lefort C, Benoist L, Chadet S, Piollet M, Heraud A, Babuty D, Baron C, Ivanes F, Angoulvant D. Stimulation of P2Y11 receptor modulates cardiac fibroblasts secretome toward immunomodulatory and protective roles after Hypoxia/Reoxygenation injury. J Mol Cell Cardiol 2018; 121:212-222. [PMID: 30031814 DOI: 10.1016/j.yjmcc.2018.07.245] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 07/10/2018] [Accepted: 07/19/2018] [Indexed: 12/21/2022]
Abstract
Cardiac fibroblasts are important regulators of myocardial structure and function. Their implications in pathological processes such as Ischemia/Reperfusion are well characterized. Cardiac fibroblasts respond to stress by excessive proliferation and secretion of pro-inflammatory cytokines and other factors, e.g. ATP, leading to purinergic receptors activation. P2Y11 receptor (P2Y11R) is an ATP-sensitive GPCR playing an immunomodulatory role in human dendritic cells (DC). We hypothesized that P2Y11R stimulation modulated the pro-inflammatory responses of human cardiac fibroblasts (HCF) to Hypoxia/Reoxygenation (H/R) mainly by acting on their secretome. P2Y11R stimulation in HCF at the onset of reoxygenation significantly limited H/R-induced proliferation (-19%) and pro-inflammatory cytokines and ATP secretion (-44% and -83% respectively). Exposure of DC to HCF secretome increased their expression of CD83, CD25 and CD86, suggesting a switch from immature to mature phenotype. Under LPS stimulation, DC had a pro-inflammatory profile (high IL-12/IL-10 ratio) that was decreased by HCF secretome (-3,8-fold), indicating induction of a tolerogenic profile. Moreover, P2Y11R inhibition in HCF led to high IL-12 secretion in DC, suggesting that the immunomodulatory effect of HCF secretome is P2Y11R-dependant. HCF secretome reduced H/R-induced cardiomyocytes death (-23%) through RISK pathway activation. P2Y11R inhibition in HCF induced a complete loss of HCF secretome protective effect, highlighting the cardioprotective role of P2Y11R. Our data demonstrated paracrine interactions between HCF, cardiomyocytes and DC following H/R, suggesting a key role of HCF in the cellular responses to reperfusion. These results also demonstrated a beneficial role of P2Y11R in HCF during H/R and strongly support the hypothesis that P2Y11R is a modulator of I/R injury.
Collapse
Affiliation(s)
- Claudie Lefort
- EA 4245 "Transplantation, Immunologie et Inflammation", Loire Valley Cardiovascular Collaboration & Université de Tours, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Lauriane Benoist
- EA 4245 "Transplantation, Immunologie et Inflammation", Loire Valley Cardiovascular Collaboration & Université de Tours, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Stéphanie Chadet
- EA 4245 "Transplantation, Immunologie et Inflammation", Loire Valley Cardiovascular Collaboration & Université de Tours, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Marie Piollet
- EA 4245 "Transplantation, Immunologie et Inflammation", Loire Valley Cardiovascular Collaboration & Université de Tours, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Audrey Heraud
- EA 4245 "Transplantation, Immunologie et Inflammation", Loire Valley Cardiovascular Collaboration & Université de Tours, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Dominique Babuty
- EA 4245 "Transplantation, Immunologie et Inflammation", Loire Valley Cardiovascular Collaboration & Université de Tours, 10 Boulevard Tonnellé, 37032 Tours, France; Service de Cardiologie, Centre Hospitalier Régional Universitaire de Tours, 37044 Tours, France
| | - Christophe Baron
- EA 4245 "Transplantation, Immunologie et Inflammation", Loire Valley Cardiovascular Collaboration & Université de Tours, 10 Boulevard Tonnellé, 37032 Tours, France; Service de Néphrologie et d'Immunologie Clinique, Centre Hospitalier Régional Universitaire de Tours, 37044 Tours, France
| | - Fabrice Ivanes
- EA 4245 "Transplantation, Immunologie et Inflammation", Loire Valley Cardiovascular Collaboration & Université de Tours, 10 Boulevard Tonnellé, 37032 Tours, France; Service de Cardiologie, Centre Hospitalier Régional Universitaire de Tours, 37044 Tours, France.
| | - Denis Angoulvant
- EA 4245 "Transplantation, Immunologie et Inflammation", Loire Valley Cardiovascular Collaboration & Université de Tours, 10 Boulevard Tonnellé, 37032 Tours, France; Service de Cardiologie, Centre Hospitalier Régional Universitaire de Tours, 37044 Tours, France
| |
Collapse
|
45
|
Ayoub KF, Pothineni NVK, Rutland J, Ding Z, Mehta JL. Immunity, Inflammation, and Oxidative Stress in Heart Failure: Emerging Molecular Targets. Cardiovasc Drugs Ther 2018; 31:593-608. [PMID: 28956198 DOI: 10.1007/s10557-017-6752-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE Heart failure (HF) remains a major cause of morbidity and mortality worldwide. Although various therapies developed over the last two decades have shown improved long term outcomes in patients with established HF, there has been little progress in preventing the adverse cardiac remodeling that initiates HF. To fill the gap in treatment, current research efforts are focused on understanding novel mechanisms and signaling pathways. Immune activation, inflammation, oxidative stress, alterations in mitochondrial bioenergetics, and autophagy have been postulated as important pathophysiological events in this process. An improved understanding of these complex processes could facilitate a therapeutic shift toward molecular targets that can potentially alter the course of HF. METHODS In this review, we address the role of immunity, inflammation, and oxidative stress as well as other novel emerging concepts in the pathophysiology of HF that may have therapeutic implications. CONCLUSION Based on the experimental and clinical studies presented here, we anticipate that a better understanding of the pathophysiology of HF will open the door for new therapeutic targets. A one-size-fits-all approach may not be appropriate for all patients with HF, and further clinical trials utilizing molecular targeting in HF may result in improved outcomes.
Collapse
Affiliation(s)
- Karam F Ayoub
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Naga Venkata K Pothineni
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Joshua Rutland
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Zufeng Ding
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jawahar L Mehta
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, USA. .,Division of Cardiovascular Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, #532, Little Rock, AR, 72205, USA.
| |
Collapse
|
46
|
Cui H, Miao S, Esworthy T, Zhou X, Lee SJ, Liu C, Yu ZX, Fisher JP, Mohiuddin M, Zhang LG. 3D bioprinting for cardiovascular regeneration and pharmacology. Adv Drug Deliv Rev 2018; 132:252-269. [PMID: 30053441 PMCID: PMC6226324 DOI: 10.1016/j.addr.2018.07.014] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/22/2018] [Accepted: 07/20/2018] [Indexed: 12/18/2022]
Abstract
Cardiovascular disease (CVD) is a major cause of morbidity and mortality worldwide. Compared to traditional therapeutic strategies, three-dimensional (3D) bioprinting is one of the most advanced techniques for creating complicated cardiovascular implants with biomimetic features, which are capable of recapitulating both the native physiochemical and biomechanical characteristics of the cardiovascular system. The present review provides an overview of the cardiovascular system, as well as describes the principles of, and recent advances in, 3D bioprinting cardiovascular tissues and models. Moreover, this review will focus on the applications of 3D bioprinting technology in cardiovascular repair/regeneration and pharmacological modeling, further discussing current challenges and perspectives.
Collapse
Affiliation(s)
- Haitao Cui
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Shida Miao
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Timothy Esworthy
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Xuan Zhou
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Se-Jun Lee
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Chengyu Liu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zu-Xi Yu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742, USA
| | | | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA; Department of Electrical and Computer Engineering, The George Washington University, Washington, DC 20052, USA; Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; Department of Medicine, The George Washington University, Washington, DC 20052, USA.
| |
Collapse
|
47
|
Trial J, Cieslik KA. Changes in cardiac resident fibroblast physiology and phenotype in aging. Am J Physiol Heart Circ Physiol 2018; 315:H745-H755. [PMID: 29906228 DOI: 10.1152/ajpheart.00237.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The cardiac fibroblast plays a central role in tissue homeostasis and in repair after injury. With aging, dysregulated cardiac fibroblasts have a reduced capacity to activate a canonical transforming growth factor-β-Smad pathway and differentiate poorly into contractile myofibroblasts. That results in the formation of an insufficient scar after myocardial infarction. In contrast, in the uninjured aged heart, fibroblasts are activated and acquire a profibrotic phenotype that leads to interstitial fibrosis, ventricular stiffness, and diastolic dysfunction, all conditions that may lead to heart failure. There is an apparent paradox in aging, wherein reparative fibrosis is impaired but interstitial, adverse fibrosis is augmented. This could be explained by analyzing the effectiveness of signaling pathways in resident fibroblasts from young versus aged hearts. Whereas defective signaling by transforming growth factor-β leads to insufficient scar formation by myofibroblasts, enhanced activation of the ERK1/2 pathway may be responsible for interstitial fibrosis mediated by activated fibroblasts. Listen to this article's corresponding podcast at https://ajpheart.podbean.com/e/fibroblast-phenotypic-changes-in-the-aging-heart/ .
Collapse
Affiliation(s)
- JoAnn Trial
- Division of Cardiovascular Sciences, Department of Medicine, Baylor College of Medicine , Houston, Texas
| | - Katarzyna A Cieslik
- Division of Cardiovascular Sciences, Department of Medicine, Baylor College of Medicine , Houston, Texas
| |
Collapse
|
48
|
Masuda S, Matsuura K, Shimizu T. Inhibition of LYPD1 is critical for endothelial network formation in bioengineered tissue with human cardiac fibroblasts. Biomaterials 2018; 166:109-121. [DOI: 10.1016/j.biomaterials.2018.03.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/26/2018] [Accepted: 03/02/2018] [Indexed: 12/23/2022]
|
49
|
Exosomes and cardiovascular cell–cell communication. Essays Biochem 2018; 62:193-204. [DOI: 10.1042/ebc20170081] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 03/26/2018] [Accepted: 03/29/2018] [Indexed: 12/21/2022]
Abstract
Exosomes have become an important player in intercellular signaling. These lipid microvesicles can stably transfer miRNA, protein, and other molecules between cells and circulate throughout the body. Exosomes are released by almost all cell types and are present in most if not all biological fluids. The biologically active cargo carried by exosomes can alter the phenotype of recipient cells. Exosomes increasingly are recognized as having an important role in the progression and treatment of cardiac disease states. Injured cardiac cells can release exosomes with important pathological effects on surrounding tissue, in addition to effecting other organs. But of equal interest is the possible benefit(s) conferred by exosomes released from stem cells for use in treatment and possible repair of cardiac damage.
Collapse
|
50
|
Baudequin T, Tabrizian M. Multilineage Constructs for Scaffold-Based Tissue Engineering: A Review of Tissue-Specific Challenges. Adv Healthc Mater 2018; 7. [PMID: 29193897 DOI: 10.1002/adhm.201700734] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/28/2017] [Indexed: 12/11/2022]
Abstract
There is a growing interest in the regeneration of tissue in interfacial regions, where biological, physical, and chemical attributes vary across tissue type. The simultaneous use of distinct cell lineages can help in developing in vitro structures, analogous to native composite tissues. This literature review gathers the recent reports that have investigated multiple cell types of various sources and lineages in a coculture system for tissue-engineered constructs. Such studies aim at mimicking the native organization of tissues and their interfaces, and/or to improve the development of complex tissue substitutes. This paper thus distinguishes itself from those focusing on technical aspects of coculturing for a single specific tissue. The first part of this review is dedicated to variables of cocultured tissue engineering such as scaffold, cells, and in vitro culture environment. Next, tissue-specific coculture methods and approaches are covered for the most studied tissues. Finally, cross-analysis is performed to highlight emerging trends in coculture principles and to discuss how tissue-specific challenges can inspire new approaches for regeneration of different interfaces to improve the outcomes of various tissue engineering strategies.
Collapse
Affiliation(s)
- Timothée Baudequin
- Faculty of Medicine; Biomat'X Laboratory; Department of Biomedical Engineering; McGill University; 740 ave. Dr. Penfield, Room 4300 Montréal QC H3A 0G1 Québec Canada
| | - Maryam Tabrizian
- Faculty of Medicine; Biomat'X Laboratory; Department of Biomedical Engineering; McGill University; 740 ave. Dr. Penfield, Room 4300 Montréal QC H3A 0G1 Québec Canada
- Faculty of Dentistry; McGill University; 3775 rue University, Room 313/308B Montréal QC H3A 2B4 Québec Canada
| |
Collapse
|