1
|
Bolesani E, Bornhorst D, Iyer LM, Zawada D, Friese N, Morgan M, Lange L, Gonzalez DM, Schrode N, Leffler A, Wunder J, Franke A, Drakhlis L, Sebra R, Schambach A, Goedel A, Dubois NC, Dobreva G, Moretti A, Zelaráyan LC, Abdelilah-Seyfried S, Zweigerdt R. Transient stabilization of human cardiovascular progenitor cells from human pluripotent stem cells in vitro reflects stage-specific heart development in vivo. Cardiovasc Res 2024; 120:1295-1311. [PMID: 38836637 DOI: 10.1093/cvr/cvae118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/11/2024] [Accepted: 04/06/2024] [Indexed: 06/06/2024] Open
Abstract
AIMS Understanding the molecular identity of human pluripotent stem cell (hPSC)-derived cardiac progenitors and mechanisms controlling their proliferation and differentiation is valuable for developmental biology and regenerative medicine. METHODS AND RESULTS Here, we show that chemical modulation of histone acetyl transferases (by IQ-1) and WNT (by CHIR99021) synergistically enables the transient and reversible block of directed cardiac differentiation progression on hPSCs. The resulting stabilized cardiovascular progenitors (SCPs) are characterized by ISL1pos/KI-67pos/NKX2-5neg expression. In the presence of the chemical inhibitors, SCPs maintain a proliferation quiescent state. Upon small molecules, removal SCPs resume proliferation and concomitant NKX2-5 up-regulation triggers cell-autonomous differentiation into cardiomyocytes. Directed differentiation of SCPs into the endothelial and smooth muscle lineages confirms their full developmental potential typical of bona fide cardiovascular progenitors. Single-cell RNA-sequencing-based transcriptional profiling of our in vitro generated human SCPs notably reflects the dynamic cellular composition of E8.25-E9.25 posterior second heart field of mouse hearts, hallmarked by nuclear receptor sub-family 2 group F member 2 expression. Investigating molecular mechanisms of SCP stabilization, we found that the cell-autonomously regulated retinoic acid and BMP signalling is governing SCP transition from quiescence towards proliferation and cell-autonomous differentiation, reminiscent of a niche-like behaviour. CONCLUSION The chemically defined and reversible nature of our stabilization approach provides an unprecedented opportunity to dissect mechanisms of cardiovascular progenitors' specification and reveal their cellular and molecular properties.
Collapse
Affiliation(s)
- Emiliano Bolesani
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Dorothee Bornhorst
- Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
- Institute of Biochemistry and Biology, Potsdam University, Potsdam, Germany
| | - Lavanya M Iyer
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
- Epigenetic Regulation and Chromatin Architecture Group, Berlin Institute for Medical Systems Biology, Max-Delbrück Centre for Molecular Medicine, Berlin, Germany
| | - Dorota Zawada
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Nina Friese
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Michael Morgan
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Lucas Lange
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - David M Gonzalez
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Nadine Schrode
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Andreas Leffler
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Julian Wunder
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Annika Franke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Lika Drakhlis
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Robert Sebra
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Alexander Goedel
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Nicole C Dubois
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Gergana Dobreva
- Department of Anatomy and Developmental Biology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alessandra Moretti
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Laura C Zelaráyan
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
| | - Salim Abdelilah-Seyfried
- Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
- Institute of Biochemistry and Biology, Potsdam University, Potsdam, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| |
Collapse
|
2
|
Boulgakoff L, Sturny R, Olejnickova V, Sedmera D, Kelly RG, Miquerol L. Participation of ventricular trabeculae in neonatal cardiac regeneration leads to ectopic recruitment of Purkinje-like cells. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1140-1157. [PMID: 39198628 DOI: 10.1038/s44161-024-00530-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 07/30/2024] [Indexed: 09/01/2024]
Abstract
Unlike adult mammals, newborn mice can regenerate a functional heart after myocardial infarction; however, the precise origin of the newly formed cardiomyocytes and whether the distal part of the conduction system (the Purkinje fiber (PF) network) is properly formed in regenerated hearts remains unclear. PFs, as well as subendocardial contractile cardiomyocytes, are derived from trabeculae, transient myocardial ridges on the inner ventricular surface. Here, using connexin 40-driven genetic tracing, we uncover a substantial participation of the trabecular lineage in myocardial regeneration through dedifferentiation and proliferation. Concomitantly, regeneration disrupted PF network maturation, resulting in permanent PF hyperplasia and impaired ventricular conduction. Proliferation assays, genetic impairment of PF recruitment, lineage tracing and clonal analysis revealed that PF network hyperplasia results from excessive recruitment of PFs due to increased trabecular fate plasticity. These data indicate that PF network hyperplasia is a consequence of trabeculae participation in myocardial regeneration.
Collapse
Affiliation(s)
- Lucie Boulgakoff
- Aix-Marseille Université, CNRS UMR 7288, Developmental Biology Institute of Marseille, Marseille, France
| | - Rachel Sturny
- Aix-Marseille Université, CNRS UMR 7288, Developmental Biology Institute of Marseille, Marseille, France
| | - Veronika Olejnickova
- Charles University, First Faculty of Medicine, Institute of Anatomy, Prague, Czech Republic
| | - David Sedmera
- Charles University, First Faculty of Medicine, Institute of Anatomy, Prague, Czech Republic
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, Developmental Biology Institute of Marseille, Marseille, France
| | - Lucile Miquerol
- Aix-Marseille Université, CNRS UMR 7288, Developmental Biology Institute of Marseille, Marseille, France.
| |
Collapse
|
3
|
Shoji JY, Davis RP, Mummery CL, Krauss S. Global Literature Analysis of Organoid and Organ-on-Chip Research. Adv Healthc Mater 2024; 13:e2301067. [PMID: 37479227 DOI: 10.1002/adhm.202301067] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/14/2023] [Indexed: 07/23/2023]
Abstract
Organoids and cells in organ-on-chip platforms replicate higher-level anatomical, physiological, or pathological states of tissues and organs. These technologies are widely regarded by academia, the pharmacological industry and regulators as key biomedical developments. To map advances in this emerging field, a literature analysis of 16,000 article metadata based on a quality-controlled text-mining algorithm is performed. The analysis covers titles, keywords, and abstracts of categorized academic publications in the literature and preprint databases published after 2010. The algorithm identifies and tracks 149 and 107 organs or organ substructures modeled as organoids and organ-on-chip, respectively, stem cell sources, as well as 130 diseases, and 16 groups of organisms other than human and mouse in which organoid/organ-on-chip technology is applied. The analysis illustrates changing diversity and focus in organoid/organ-on-chip research and captures its geographical distribution. The downloadable dataset provided is a robust framework for researchers to interrogate with their own questions.
Collapse
Affiliation(s)
- Jun-Ya Shoji
- Hybrid Technology Hub, Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, 0372, Norway
| | - Richard P Davis
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, 2300RC, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, 2300RC, the Netherlands
| | - Christine L Mummery
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, 2300RC, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, 2300RC, the Netherlands
- Department of Applied Stem Cell Technologies, University of Twente, Enschede, 7522NB, the Netherlands
| | - Stefan Krauss
- Hybrid Technology Hub, Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, 0372, Norway
| |
Collapse
|
4
|
Yang X, He S, Li X, Guo Z, Wang H, Zhang Z, Song X, Jia K, He L, Zhou B. Synchronized lineage tracing of cell membranes and nuclei by dual recombinases and dual fluorescent. J Genet Genomics 2024:S1673-8527(24)00182-6. [PMID: 38996840 DOI: 10.1016/j.jgg.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/14/2024]
Abstract
Genetic lineage tracing has been widely employed to investigate cell lineages and fate. However, conventional reporting systems often label the entire cytoplasm, making it challenging to discern cell boundaries. Additionally, single Cre-loxP recombination systems have limitations in tracing specific cell populations. This study proposes three reporting systems that utilizing Cre, Dre, and Dre + Cre mediated recombination. These systems incorporate tdTomato expression on the cell membrane and PhiYFP expression within the nucleus, allowing for clear observation of the nucleus and membrane. The efficacy of these systems is successfully demonstrated by labeling cardiomyocytes and hepatocytes. The potential for dynamic visualization of the cell membrane is showcased using intravital imaging microscopy or three-dimensional imaging. Furthermore, by combining this dual recombinase system with the ProTracer system, hepatocyte proliferation is traced with enhanced precision. This reporting system holds significant importance for advancing the understanding of cell fate studies in development, homeostasis, and diseases.
Collapse
Affiliation(s)
- Xueying Yang
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| | - Shun He
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Xufeng Li
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| | - Zhihou Guo
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Haichang Wang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Zhuonan Zhang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Xin Song
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Ke Jia
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Lingjuan He
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China.
| | - Bin Zhou
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China; New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
5
|
Groen E, Mummery CL, Yiangou L, Davis RP. Three-dimensional cardiac models: a pre-clinical testing platform. Biochem Soc Trans 2024; 52:1045-1059. [PMID: 38778769 PMCID: PMC11346450 DOI: 10.1042/bst20230444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/25/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
Major advancements in human pluripotent stem cell (hPSC) technology over recent years have yielded valuable tools for cardiovascular research. Multi-cell type 3-dimensional (3D) cardiac models in particular, are providing complementary approaches to animal studies that are better representatives than simple 2-dimensional (2D) cultures of differentiated hPSCs. These human 3D cardiac models can be broadly divided into two categories; namely those generated through aggregating pre-differentiated cells and those that form self-organizing structures during their in vitro differentiation from hPSCs. These models can either replicate aspects of cardiac development or enable the examination of interactions among constituent cell types, with some of these models showing increased maturity compared with 2D systems. Both groups have already emerged as physiologically relevant pre-clinical platforms for studying heart disease mechanisms, exhibiting key functional attributes of the human heart. In this review, we describe the different cardiac organoid models derived from hPSCs, their generation methods, applications in cardiovascular disease research and use in drug screening. We also address their current limitations and challenges as pre-clinical testing platforms and propose potential improvements to enhance their efficacy in cardiac drug discovery.
Collapse
Affiliation(s)
- Eline Groen
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - Loukia Yiangou
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Richard P. Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, 2300RC Leiden, The Netherlands
| |
Collapse
|
6
|
Palmer JA, Rosenthal N, Teichmann SA, Litvinukova M. Revisiting Cardiac Biology in the Era of Single Cell and Spatial Omics. Circ Res 2024; 134:1681-1702. [PMID: 38843288 PMCID: PMC11149945 DOI: 10.1161/circresaha.124.323672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 06/09/2024]
Abstract
Throughout our lifetime, each beat of the heart requires the coordinated action of multiple cardiac cell types. Understanding cardiac cell biology, its intricate microenvironments, and the mechanisms that govern their function in health and disease are crucial to designing novel therapeutical and behavioral interventions. Recent advances in single-cell and spatial omics technologies have significantly propelled this understanding, offering novel insights into the cellular diversity and function and the complex interactions of cardiac tissue. This review provides a comprehensive overview of the cellular landscape of the heart, bridging the gap between suspension-based and emerging in situ approaches, focusing on the experimental and computational challenges, comparative analyses of mouse and human cardiac systems, and the rising contextualization of cardiac cells within their niches. As we explore the heart at this unprecedented resolution, integrating insights from both mouse and human studies will pave the way for novel diagnostic tools and therapeutic interventions, ultimately improving outcomes for patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Jack A. Palmer
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom (J.A.P., S.A.T.)
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus (J.A.P., S.A.T.), University of Cambridge, United Kingdom
| | - Nadia Rosenthal
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME (N.R.)
- National Heart and Lung Institute, Imperial College London, United Kingdom (N.R.)
| | - Sarah A. Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom (J.A.P., S.A.T.)
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus (J.A.P., S.A.T.), University of Cambridge, United Kingdom
- Theory of Condensed Matter Group, Department of Physics, Cavendish Laboratory (S.A.T.), University of Cambridge, United Kingdom
| | - Monika Litvinukova
- University Hospital Würzburg, Germany (M.L.)
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, Germany (M.L.)
- Helmholtz Pioneer Campus, Helmholtz Munich, Germany (M.L.)
| |
Collapse
|
7
|
Zhu C, Yuan T, Krishnan J. Targeting cardiomyocyte cell cycle regulation in heart failure. Basic Res Cardiol 2024; 119:349-369. [PMID: 38683371 PMCID: PMC11142990 DOI: 10.1007/s00395-024-01049-x] [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: 11/21/2023] [Revised: 03/11/2024] [Accepted: 03/29/2024] [Indexed: 05/01/2024]
Abstract
Heart failure continues to be a significant global health concern, causing substantial morbidity and mortality. The limited ability of the adult heart to regenerate has posed challenges in finding effective treatments for cardiac pathologies. While various medications and surgical interventions have been used to improve cardiac function, they are not able to address the extensive loss of functioning cardiomyocytes that occurs during cardiac injury. As a result, there is growing interest in understanding how the cell cycle is regulated and exploring the potential for stimulating cardiomyocyte proliferation as a means of promoting heart regeneration. This review aims to provide an overview of current knowledge on cell cycle regulation and mechanisms underlying cardiomyocyte proliferation in cases of heart failure, while also highlighting established and novel therapeutic strategies targeting this area for treatment purposes.
Collapse
Affiliation(s)
- Chaonan Zhu
- Department of Medicine III, Cardiology/Angiology/Nephrology, Goethe University Hospital, 60590, Frankfurt am Main, Germany
- Institute for Cardiovascular Regeneration, Goethe University, 60590, Frankfurt am Main, Germany
| | - Ting Yuan
- Department of Medicine III, Cardiology/Angiology/Nephrology, Goethe University Hospital, 60590, Frankfurt am Main, Germany.
- Institute for Cardiovascular Regeneration, Goethe University, 60590, Frankfurt am Main, Germany.
- German Center for Cardiovascular Research, Partner Site Rhein-Main, 60590, Frankfurt am Main, Germany.
- Cardio-Pulmonary Institute, Goethe University Hospital, 60590, Frankfurt am Main, Germany.
| | - Jaya Krishnan
- Department of Medicine III, Cardiology/Angiology/Nephrology, Goethe University Hospital, 60590, Frankfurt am Main, Germany.
- Institute for Cardiovascular Regeneration, Goethe University, 60590, Frankfurt am Main, Germany.
- German Center for Cardiovascular Research, Partner Site Rhein-Main, 60590, Frankfurt am Main, Germany.
- Cardio-Pulmonary Institute, Goethe University Hospital, 60590, Frankfurt am Main, Germany.
| |
Collapse
|
8
|
Torimoto K, Elliott K, Nakayama Y, Yanagisawa H, Eguchi S. Cardiac and perivascular myofibroblasts, matrifibrocytes, and immune fibrocytes in hypertension; commonalities and differences with other cardiovascular diseases. Cardiovasc Res 2024; 120:567-580. [PMID: 38395029 PMCID: PMC11485269 DOI: 10.1093/cvr/cvae044] [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: 09/18/2023] [Revised: 02/02/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Hypertension is a major cause of cardiovascular diseases such as myocardial infarction and stroke. Cardiovascular fibrosis occurs with hypertension and contributes to vascular resistance, aortic stiffness, and cardiac hypertrophy. However, the molecular mechanisms leading to fibroblast activation in hypertension remain largely unknown. There are two types of fibrosis: replacement fibrosis and reactive fibrosis. Replacement fibrosis occurs in response to the loss of viable tissue to form a scar. Reactive fibrosis occurs in response to an increase in mechanical and neurohormonal stress. Although both types of fibrosis are considered adaptive processes, they become maladaptive when the tissue loss is too large, or the stress persists. Myofibroblasts represent a subpopulation of activated fibroblasts that have gained contractile function to promote wound healing. Therefore, myofibroblasts are a critical cell type that promotes replacement fibrosis. Although myofibroblasts were recognized as the fibroblasts participating in reactive fibrosis, recent experimental evidence indicated there are distinct fibroblast populations in cardiovascular reactive fibrosis. Accordingly, we will discuss the updated definition of fibroblast subpopulations, the regulatory mechanisms, and their potential roles in cardiovascular pathophysiology utilizing new knowledge from various lineage tracing and single-cell RNA sequencing studies. Among the fibroblast subpopulations, we will highlight the novel roles of matrifibrocytes and immune fibrocytes in cardiovascular fibrosis including experimental models of hypertension, pressure overload, myocardial infarction, atherosclerosis, aortic aneurysm, and nephrosclerosis. Exploration into the molecular mechanisms involved in the differentiation and activation of those fibroblast subpopulations may lead to novel treatments for end-organ damage associated with hypertension and other cardiovascular diseases.
Collapse
Affiliation(s)
- Keiichi Torimoto
- Department of Cardiovascular Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Katherine Elliott
- Department of Cardiovascular Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Yuki Nakayama
- Department of Cardiovascular Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Hiromi Yanagisawa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
- Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Satoru Eguchi
- Department of Cardiovascular Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
9
|
Jiang Y, Wei ZY, Song ZF, Qian HY. Platelet-inspired targeting delivery for coronary heart disease. Heliyon 2024; 10:e27166. [PMID: 38449604 PMCID: PMC10915553 DOI: 10.1016/j.heliyon.2024.e27166] [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: 10/03/2023] [Revised: 01/08/2024] [Accepted: 02/26/2024] [Indexed: 03/08/2024] Open
Abstract
Platelets play a pivotal role in many physiological and pathological processes, with their special targeting/adhering properties towards infarcted myocardium, injured or dysfunctional endothelium, and growing thrombus. Leveraging the site-targeting/adhering property, a variety of platelet-inspired targeting delivery(PITD)designs have been developed, the majority of which are reached by hitchhiking live platelets, cloaking nanoparticles with platelet membranes and mimicking platelet functions. With PITD, drugs or regenerative cells can directly reach targeted sites with minimized systematical distribution thus being of great clinical benefits. Coronary heart disease (CHD) is a major health burden worldwide. Plenty of PITD designs have shown promising outcomes for the treatment of CHD in preclinical models, especially in thrombolysis and post-percutaneous coronary intervention (post-PCI) anti-restenosis. Besides, PITD applications in cardiac protection and atherosclerotic plaque imaging are also under investigation. What's more, the potential benefits of PITD in the field of cell-based therapy are also attracting growing attention since it may resolve the problem of low arriving and retention efficiency, which are also particularly discussed in this review. In brief, our focus is putting on PITD strategies designed for the treatment of CHD, which hopefully can facilitate further optimization of this direction.
Collapse
Affiliation(s)
| | | | | | - Hai-Yan Qian
- Center for Coronary Heart Disease, Department of Cardiology, Fu Wai Hospital, National Center for Cardiovascular Diseases of China, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
10
|
Avolio E, Campagnolo P, Katare R, Madeddu P. The role of cardiac pericytes in health and disease: therapeutic targets for myocardial infarction. Nat Rev Cardiol 2024; 21:106-118. [PMID: 37542118 DOI: 10.1038/s41569-023-00913-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2023] [Indexed: 08/06/2023]
Abstract
Millions of cardiomyocytes die immediately after myocardial infarction, regardless of whether the culprit coronary artery undergoes prompt revascularization. Residual ischaemia in the peri-infarct border zone causes further cardiomyocyte damage, resulting in a progressive decline in contractile function. To date, no treatment has succeeded in increasing the vascularization of the infarcted heart. In the past decade, new approaches that can target the heart's highly plastic perivascular niche have been proposed. The perivascular environment is populated by mesenchymal progenitor cells, fibroblasts, myofibroblasts and pericytes, which can together mount a healing response to the ischaemic damage. In the infarcted heart, pericytes have crucial roles in angiogenesis, scar formation and stabilization, and control of the inflammatory response. Persistent ischaemia and accrual of age-related risk factors can lead to pericyte depletion and dysfunction. In this Review, we describe the phenotypic changes that characterize the response of cardiac pericytes to ischaemia and the potential of pericyte-based therapy for restoring the perivascular niche after myocardial infarction. Pericyte-related therapies that can salvage the area at risk of an ischaemic injury include exogenously administered pericytes, pericyte-derived exosomes, pericyte-engineered biomaterials, and pharmacological approaches that can stimulate the differentiation of constitutively resident pericytes towards an arteriogenic phenotype. Promising preclinical results from in vitro and in vivo studies indicate that pericytes have crucial roles in the treatment of coronary artery disease and the prevention of post-ischaemic heart failure.
Collapse
Affiliation(s)
- Elisa Avolio
- Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol, UK.
| | - Paola Campagnolo
- School of Biosciences, Faculty of Health & Medical Sciences, University of Surrey, Guildford, UK
| | - Rajesh Katare
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Paolo Madeddu
- Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol, UK.
| |
Collapse
|
11
|
Weinberger M, Riley PR. Animal models to study cardiac regeneration. Nat Rev Cardiol 2024; 21:89-105. [PMID: 37580429 DOI: 10.1038/s41569-023-00914-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/17/2023] [Indexed: 08/16/2023]
Abstract
Permanent fibrosis and chronic deterioration of heart function in patients after myocardial infarction present a major health-care burden worldwide. In contrast to the restricted potential for cellular and functional regeneration of the adult mammalian heart, a robust capacity for cardiac regeneration is seen during the neonatal period in mammals as well as in the adults of many fish and amphibian species. However, we lack a complete understanding as to why cardiac regeneration takes place more efficiently in some species than in others. The capacity of the heart to regenerate after injury is controlled by a complex network of cellular and molecular mechanisms that form a regulatory landscape, either permitting or restricting regeneration. In this Review, we provide an overview of the diverse array of vertebrates that have been studied for their cardiac regenerative potential and discuss differential heart regeneration outcomes in closely related species. Additionally, we summarize current knowledge about the core mechanisms that regulate cardiac regeneration across vertebrate species.
Collapse
Affiliation(s)
- Michael Weinberger
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Paul R Riley
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
12
|
Li Y, Liu Z, Han X, Liang F, Zhang Q, Huang X, Shi X, Huo H, Han M, Liu X, Zhu H, He L, Shen L, Hu X, Wang J, Wang QD, Smart N, Zhou B, He B. Dynamics of Endothelial Cell Generation and Turnover in Arteries During Homeostasis and Diseases. Circulation 2024; 149:135-154. [PMID: 38084582 DOI: 10.1161/circulationaha.123.064301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 10/06/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND Endothelial cell (EC) generation and turnover by self-proliferation contributes to vascular repair and regeneration. The ability to accurately measure the dynamics of EC generation would advance our understanding of cellular mechanisms of vascular homeostasis and diseases. However, it is currently challenging to evaluate the dynamics of EC generation in large vessels such as arteries because of their infrequent proliferation. METHODS By using dual recombination systems based on Cre-loxP and Dre-rox, we developed a genetic system for temporally seamless recording of EC proliferation in vivo. We combined genetic recording of EC proliferation with single-cell RNA sequencing and gene knockout to uncover cellular and molecular mechanisms underlying EC generation in arteries during homeostasis and disease. RESULTS Genetic proliferation tracing reveals that ≈3% of aortic ECs undergo proliferation per month in adult mice during homeostasis. The orientation of aortic EC division is generally parallel to blood flow in the aorta, which is regulated by the mechanosensing protein Piezo1. Single-cell RNA sequencing analysis reveals 4 heterogeneous aortic EC subpopulations with distinct proliferative activity. EC cluster 1 exhibits transit-amplifying cell features with preferential proliferative capacity and enriched expression of stem cell markers such as Sca1 and Sox18. EC proliferation increases in hypertension but decreases in type 2 diabetes, coinciding with changes in the extent of EC cluster 1 proliferation. Combined gene knockout and proliferation tracing reveals that Hippo/vascular endothelial growth factor receptor 2 signaling pathways regulate EC proliferation in large vessels. CONCLUSIONS Genetic proliferation tracing quantitatively delineates the dynamics of EC generation and turnover, as well as EC division orientation, in large vessels during homeostasis and disease. An EC subpopulation in the aorta exhibits more robust cell proliferation during homeostasis and type 2 diabetes, identifying it as a potential therapeutic target for vascular repair and regeneration.
Collapse
Affiliation(s)
- Yi Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Zixin Liu
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Ximeng Han
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Feng Liang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Qianyu Zhang
- School of Life Science and Technology, ShanghaiTech University, China (Q.Z., M.H., B.Z.)
| | - Xiuzhen Huang
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Xin Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Huanhuan Huo
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Maoying Han
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
- School of Life Science and Technology, ShanghaiTech University, China (Q.Z., M.H., B.Z.)
| | - Xiuxiu Liu
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Huan Zhu
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Lingjuan He
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China (L.H.)
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Xinyang Hu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (X.H., J.W.)
| | - Jian'an Wang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (X.H., J.W.)
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (Q.D.W.)
| | - Nicola Smart
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, UK (N.S.)
| | - Bin Zhou
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
- School of Life Science and Technology, ShanghaiTech University, China (Q.Z., M.H., B.Z.)
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, China (B.Z.)
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| |
Collapse
|
13
|
Hénon P, Bischoff N, Dallemand R. Transforming Perspectives in Cardiac Cell Therapy: Hypothesis and Commentary Following Updated Results of a Pilot Study Investigating Very Long-Term Clinical Outcomes in Severe AMI Patients Following Trans-Epicardial Injection of Peripheral Blood CD34 + Cells. Stem Cell Rev Rep 2024; 20:247-257. [PMID: 37861968 PMCID: PMC10799833 DOI: 10.1007/s12015-023-10643-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2023] [Indexed: 10/21/2023]
Abstract
Ischemic heart attack is the leading cause of death worldwide. Ten percent of cases will die within an hour. Of the survivors, around 30% will have suffered a severe infarction which will lead to the irreparable destruction of 1 to 2 billion myocardial cells, causing an irreversible secondary heart failure with a poor prognosis in the short. The heart is a totally differentiated organ with a very low capacity for self-regeneration. No current treatment can prevent this fatal outcome, but only slow it down. For these reasons, cell therapy has generated enormous hope, but achieved somewhat disappointing results, depending on the type/source of cells which were used. From the end of 2002, our group conducted a Pilot study using immuno-selected autologous peripheral-blood (PB) CD34+ cells in a small cohort of patients who had experienced a heart attack with poor prognosis. Three of these patients were immediately considered for heart transplant but lacked a readily available donor. CD34+ cells were trans-epicardially delivered at the end of a coronary artery by-pass graft (CABG) operation without reperfusing the ischemic area, which was performed on a compassionate basis. All but one patient showed a marked and sustained improvement in their cardiac function parameters from the baseline values, associated with both cardiac tissue repair and revascularization, as demonstrated by PetScan examination. The patients' outcomes have been recently updated. Six out of seven patients have survived in good enough conditions for at least 12 years after cell therapy, including those three initially recommended for heart transplant and who have avoided it. Presently, five out of seven patients are still alive with an average follow-up of 17 years (range 16-20 years), which is very unusual after CABG for patients with such a poor initially prognosis.
Collapse
Affiliation(s)
- Philippe Hénon
- Institut de Recherche en Hématologie Et Transplantation, Hôpital du Hasenrain, 87 Avenue d'Altkirch, 68100, Mulhouse, France.
- CellProthera SAS, 12 Rue du Parc, 68100, Mulhouse, France.
| | - Nicolas Bischoff
- Département de Chirurgie Cardio-Thoracique, Groupe Hospitalier Régional Mulhouse Sud-Alsace, 20 Rue du Docteur Laënnec, 68100, Mulhouse, France
| | - Robert Dallemand
- Département de Chirurgie Cardio-Thoracique, Groupe Hospitalier Régional Mulhouse Sud-Alsace, 20 Rue du Docteur Laënnec, 68100, Mulhouse, France
| |
Collapse
|
14
|
Hardy SA, Liesinger L, Patrick R, Poettler M, Rech L, Gindlhuber J, Mabotuwana NS, Ashour D, Stangl V, Bigland M, Murtha LA, Starkey MR, Scherr D, Hansbro PM, Hoefler G, Campos Ramos G, Cochain C, Harvey RP, Birner-Gruenberger R, Boyle AJ, Rainer PP. Extracellular Matrix Protein-1 as a Mediator of Inflammation-Induced Fibrosis After Myocardial Infarction. JACC Basic Transl Sci 2023; 8:1539-1554. [PMID: 38205347 PMCID: PMC10774582 DOI: 10.1016/j.jacbts.2023.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 05/17/2023] [Accepted: 05/17/2023] [Indexed: 01/12/2024]
Abstract
Irreversible fibrosis is a hallmark of myocardial infarction (MI) and heart failure. Extracellular matrix protein-1 (ECM-1) is up-regulated in these hearts, localized to fibrotic, inflammatory, and perivascular areas. ECM-1 originates predominantly from fibroblasts, macrophages, and pericytes/vascular cells in uninjured human and mouse hearts, and from M1 and M2 macrophages and myofibroblasts after MI. ECM-1 stimulates fibroblast-to-myofibroblast transition, up-regulates key fibrotic and inflammatory pathways, and inhibits cardiac fibroblast migration. ECM-1 binds HuCFb cell surface receptor LRP1, and LRP1 inhibition blocks ECM-1 from stimulating fibroblast-to-myofibroblast transition, confirming a novel ECM-1-LRP1 fibrotic signaling axis. ECM-1 may represent a novel mechanism facilitating inflammation-fibrosis crosstalk.
Collapse
Affiliation(s)
- Sean A. Hardy
- Department of Internal Medicine and University Heart Center, Division of Cardiology, Medical University of Graz, Graz, Austria
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Laura Liesinger
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
- Institute of Chemical Technologies and Analytical Chemistry, Technische Universität Wien, Vienna, Austria
| | - Ralph Patrick
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Maria Poettler
- Department of Internal Medicine and University Heart Center, Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Lavinia Rech
- Department of Internal Medicine and University Heart Center, Division of Cardiology, Medical University of Graz, Graz, Austria
- Department of Cardiac Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Nishani S. Mabotuwana
- Department of Internal Medicine and University Heart Center, Division of Cardiology, Medical University of Graz, Graz, Austria
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - DiyaaEldin Ashour
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Verena Stangl
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Mark Bigland
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Lucy A. Murtha
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Malcolm R. Starkey
- Department of Immunology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Daniel Scherr
- Department of Internal Medicine and University Heart Center, Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Philip M. Hansbro
- Centre for Inflammation, Centenary Institute, and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, New South Wales, Australia
| | - Gerald Hoefler
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Gustavo Campos Ramos
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
- Department of Internal Medicine 1, University Hospital of Würzburg, Würzburg, Germany
| | - Clement Cochain
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Richard P. Harvey
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, UNSW Sydney, Sydney, Australia
| | - Ruth Birner-Gruenberger
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
- Institute of Chemical Technologies and Analytical Chemistry, Technische Universität Wien, Vienna, Austria
- BioTechMed Graz, Graz, Austria
| | - Andrew J. Boyle
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Department of Cardiovascular Medicine, John Hunter Hospital, New Lambton Heights, New South Wales, Australia
| | - Peter P. Rainer
- Department of Internal Medicine and University Heart Center, Division of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Department of Medicine, St. Johann in Tirol General Hospital, St. Johann in Tirol, Austria
| |
Collapse
|
15
|
Berkeley B, Tang MNH, Brittan M. Mechanisms regulating vascular and lymphatic regeneration in the heart after myocardial infarction. J Pathol 2023; 260:666-678. [PMID: 37272582 PMCID: PMC10953458 DOI: 10.1002/path.6093] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/14/2023] [Accepted: 04/27/2023] [Indexed: 06/06/2023]
Abstract
Myocardial infarction, caused by a thrombus or coronary vascular occlusion, leads to irreversible ischaemic injury. Advances in early reperfusion strategies have significantly reduced short-term mortality after myocardial infarction. However, survivors have an increased risk of developing heart failure, which confers a high risk of death at 1 year. The capacity of the injured neonatal mammalian heart to regenerate has stimulated extensive research into whether recapitulation of developmental regeneration programmes may be beneficial in adult cardiovascular disease. Restoration of functional blood and lymphatic vascular networks in the infarct and border regions via neovascularisation and lymphangiogenesis, respectively, is a key requirement to facilitate myocardial regeneration. An improved understanding of the endogenous mechanisms regulating coronary vascular and lymphatic expansion and function in development and in adult patients after myocardial infarction may inform future therapeutic strategies and improve translation from pre-clinical studies. In this review, we explore the underpinning research and key findings in the field of cardiovascular regeneration, with a focus on neovascularisation and lymphangiogenesis, and discuss the outcomes of therapeutic strategies employed to date. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Bronwyn Berkeley
- Centre for Cardiovascular Science, The Queen's Medical Research InstituteUniversity of EdinburghEdinburghUK
| | - Michelle Nga Huen Tang
- Centre for Cardiovascular Science, The Queen's Medical Research InstituteUniversity of EdinburghEdinburghUK
| | - Mairi Brittan
- Centre for Cardiovascular Science, The Queen's Medical Research InstituteUniversity of EdinburghEdinburghUK
| |
Collapse
|
16
|
Hammond T, Sage J. Monitoring the Cell Cycle of Tumor Cells in Mouse Models of Human Cancer. Cold Spring Harb Perspect Med 2023; 13:a041383. [PMID: 37460156 PMCID: PMC10691483 DOI: 10.1101/cshperspect.a041383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Cell division is obligatory to tumor growth. However, both cancer cells and noncancer cells in tumors can be found in distinct stages of the cell cycle, which may inform the growth potential of these tumors, their propensity to metastasize, and their response to therapy. Hence, it is of utmost importance to monitor the cell cycle of tumor cells. Here we discuss well-established methods and new genetic advances to track the cell cycle of tumor cells in mouse models of human cancer. We also review recent genetic studies investigating the role of the cell-cycle machinery in the growth of tumors in vivo, with a focus on the machinery regulating the G1/S transition of the cell cycle.
Collapse
Affiliation(s)
- Taylar Hammond
- Department of Pediatrics, Stanford University, Stanford, California 94305, USA
- Department of Biology, and Stanford University, Stanford, California 94305, USA
| | - Julien Sage
- Department of Pediatrics, Stanford University, Stanford, California 94305, USA
- Department of Genetics, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
17
|
Liu X, Weng W, He L, Zhou B. Genetic recording of in vivo cell proliferation by ProTracer. Nat Protoc 2023:10.1038/s41596-023-00833-8. [PMID: 37268780 DOI: 10.1038/s41596-023-00833-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 03/17/2023] [Indexed: 06/04/2023]
Abstract
The ability to experimentally measure cell proliferation is the basis for understanding the sources of cells that drive organ development, tissue regeneration and repair. Recently, we generated a genetic approach to detect cell proliferation: we used genetic lineage-tracing technologies to achieve seamless recording of in vivo cell proliferation in a tissue-specific manner. We provide a detailed protocol (generation of mouse lines, characterization of mouse lines, mouse line crossing and cell-proliferation tracing) for using this genetic system to study cell proliferation. This cell-proliferation tracing system, which we term 'ProTracer' (Proliferation Tracer), permits lifelong noninvasive monitoring of cell proliferation of specific cell lineages in live animals. Compared with other short-term strategies that require execution of animals, ProTracer does not require sampling or animal sacrifice for tissue processing. To highlight these features, we used ProTracer to study the proliferation of hepatocytes during liver homeostasis and after tissue injury in mice. We show that the protocol is applicable to study any in vivo cell proliferation, which takes ~9 months to finish from mouse generation to data analysis. This protocol can easily be carried out by researchers skilled in mouse-related experiments.
Collapse
Affiliation(s)
- Xiuxiu Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Wendong Weng
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Lingjuan He
- School of Life Sciences, Westlake University, Hangzhou, China.
| | - Bin Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
- New Cornerstone Science Laboratory, Shenzhen, China.
| |
Collapse
|
18
|
Abstract
Although osteoblasts and osteocytes are descended from the same lineage, they each have unique and essential roles in bone. Targeting gene deletion to osteoblasts and osteocytes using the Cre/loxP system has greatly increased our current understanding of how these cells function. Additionally, the use of the Cre/loxP system in conjunction with cell-specific reporters has enabled lineage tracing of these bone cells both in vivo and ex vivo. However, concerns have been raised regarding the specificity of the promoters used and the resulting off-target effects on cells within and outside of the bone. In this review, we have summarized the main mouse models that have been used to determine the functions of specific genes in osteoblasts and osteocytes. We discuss the expression patterns and specificity of the different promoter fragments during osteoblast to osteocyte differentiation in vivo. We also highlight how their expression in non-skeletal tissues may complicate the interpretation of study results. A thorough understanding of when and where these promoters are activated will enable improved study design and greater confidence in data interpretation.
Collapse
Affiliation(s)
- Y Kitase
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, United States of America
| | - M Prideaux
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, United States of America.
| |
Collapse
|
19
|
Cohen CD, Rousseau ST, Bermea KC, Bhalodia A, Lovell JP, Dina Zita M, Čiháková D, Adamo L. Myocardial Immune Cells: The Basis of Cardiac Immunology. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1198-1207. [PMID: 37068299 PMCID: PMC10111214 DOI: 10.4049/jimmunol.2200924] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/14/2023] [Indexed: 04/19/2023]
Abstract
The mammalian heart is characterized by the presence of striated myocytes, which allow continuous rhythmic contraction from early embryonic development until the last moments of life. However, the myocardium contains a significant contingent of leukocytes from every major class. This leukocyte pool includes both resident and nonresident immune cells. Over recent decades, it has become increasingly apparent that the heart is intimately sensitive to immune signaling and that myocardial leukocytes exhibit an array of critical functions, both in homeostasis and in the context of cardiac adaptation to injury. Here, we systematically review current knowledge of all major leukocyte classes in the heart, discussing their functions in health and disease. We also highlight the connection between the myocardium, immune cells, lymphoid organs, and both local and systemic immune responses.
Collapse
Affiliation(s)
- Charles D. Cohen
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Sylvie T. Rousseau
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Kevin C. Bermea
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Aashik Bhalodia
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Jana P. Lovell
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Marcelle Dina Zita
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Daniela Čiháková
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Luigi Adamo
- Cardiac Immunology Laboratory, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
20
|
Perveen S, Vanni R, Lo Iacono M, Rastaldo R, Giachino C. Direct Reprogramming of Resident Non-Myocyte Cells and Its Potential for In Vivo Cardiac Regeneration. Cells 2023; 12:1166. [PMID: 37190075 PMCID: PMC10136631 DOI: 10.3390/cells12081166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/10/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Cardiac diseases are the foremost cause of morbidity and mortality worldwide. The heart has limited regenerative potential; therefore, lost cardiac tissue cannot be replenished after cardiac injury. Conventional therapies are unable to restore functional cardiac tissue. In recent decades, much attention has been paid to regenerative medicine to overcome this issue. Direct reprogramming is a promising therapeutic approach in regenerative cardiac medicine that has the potential to provide in situ cardiac regeneration. It consists of direct cell fate conversion of one cell type into another, avoiding transition through an intermediary pluripotent state. In injured cardiac tissue, this strategy directs transdifferentiation of resident non-myocyte cells (NMCs) into mature functional cardiac cells that help to restore the native tissue. Over the years, developments in reprogramming methods have suggested that regulation of several intrinsic factors in NMCs can help to achieve in situ direct cardiac reprogramming. Among NMCs, endogenous cardiac fibroblasts have been studied for their potential to be directly reprogrammed into both induced cardiomyocytes and induced cardiac progenitor cells, while pericytes can transdifferentiate towards endothelial cells and smooth muscle cells. This strategy has been indicated to improve heart function and reduce fibrosis after cardiac injury in preclinical models. This review summarizes the recent updates and progress in direct cardiac reprogramming of resident NMCs for in situ cardiac regeneration.
Collapse
Affiliation(s)
| | - Roberto Vanni
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy
| | | | | | | |
Collapse
|
21
|
Gromova T, Gehred ND, Vondriska TM. Single-cell transcriptomes in the heart: when every epigenome counts. Cardiovasc Res 2023; 119:64-78. [PMID: 35325060 PMCID: PMC10233279 DOI: 10.1093/cvr/cvac040] [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: 10/06/2021] [Revised: 02/03/2022] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
The response of an organ to stimuli emerges from the actions of individual cells. Recent cardiac single-cell RNA-sequencing studies of development, injury, and reprogramming have uncovered heterogeneous populations even among previously well-defined cell types, raising questions about what level of experimental resolution corresponds to disease-relevant, tissue-level phenotypes. In this review, we explore the biological meaning behind this cellular heterogeneity by undertaking an exhaustive analysis of single-cell transcriptomics in the heart (including a comprehensive, annotated compendium of studies published to date) and evaluating new models for the cardiac function that have emerged from these studies (including discussion and schematics that depict new hypotheses in the field). We evaluate the evidence to support the biological actions of newly identified cell populations and debate questions related to the role of cell-to-cell variability in development and disease. Finally, we present emerging epigenomic approaches that, when combined with single-cell RNA-sequencing, can resolve basic mechanisms of gene regulation and variability in cell phenotype.
Collapse
Affiliation(s)
- Tatiana Gromova
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Medicine/Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Natalie D Gehred
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Medicine/Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Thomas M Vondriska
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Medicine/Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
22
|
Siatra P, Vatsellas G, Chatzianastasiou A, Balafas E, Manolakou T, Papapetropoulos A, Agapaki A, Mouchtouri ET, Ruchaya PJ, Korovesi AG, Mavroidis M, Thanos D, Beis D, Kokkinopoulos I. Return of the Tbx5; lineage-tracing reveals ventricular cardiomyocyte-like precursors in the injured adult mammalian heart. NPJ Regen Med 2023; 8:13. [PMID: 36869039 PMCID: PMC9984483 DOI: 10.1038/s41536-023-00280-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 01/25/2023] [Indexed: 03/05/2023] Open
Abstract
The single curative measure for heart failure patients is a heart transplantation, which is limited due to a shortage of donors, the need for immunosuppression and economic costs. Therefore, there is an urgent unmet need for identifying cell populations capable of cardiac regeneration that we will be able to trace and monitor. Injury to the adult mammalian cardiac muscle, often leads to a heart attack through the irreversible loss of a large number of cardiomyocytes, due to an idle regenerative capability. Recent reports in zebrafish indicate that Tbx5a is a vital transcription factor for cardiomyocyte regeneration. Preclinical data underscore the cardioprotective role of Tbx5 upon heart failure. Data from our earlier murine developmental studies have identified a prominent unipotent Tbx5-expressing embryonic cardiac precursor cell population able to form cardiomyocytes, in vivo, in vitro and ex vivo. Using a developmental approach to an adult heart injury model and by employing a lineage-tracing mouse model as well as the use of single-cell RNA-seq technology, we identify a Tbx5-expressing ventricular cardiomyocyte-like precursor population, in the injured adult mammalian heart. The transcriptional profile of that precursor cell population is closer to that of neonatal than embryonic cardiomyocyte precursors. Tbx5, a cardinal cardiac development transcription factor, lies in the center of a ventricular adult precursor cell population, which seems to be affected by neurohormonal spatiotemporal cues. The identification of a Tbx5-specific cardiomyocyte precursor-like cell population, which is capable of dedifferentiating and potentially deploying a cardiomyocyte regenerative program, provides a clear target cell population for translationally-relevant heart interventional studies.
Collapse
Affiliation(s)
- Panagiota Siatra
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Giannis Vatsellas
- Center for Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Greek Genome Center, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Athanasia Chatzianastasiou
- Department of Pharmacy, Laboratory of Pharmacology, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Balafas
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Theodora Manolakou
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Andreas Papapetropoulos
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Department of Pharmacy, Laboratory of Pharmacology, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Agapaki
- Histochemistry Facility, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | | | - Prashant J Ruchaya
- School of Health, Sport and Biosciences, University of East London, London, UK
| | - Artemis G Korovesi
- Center for Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Greek Genome Center, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Manolis Mavroidis
- Center for Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Dimitrios Thanos
- Center for Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Greek Genome Center, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Dimitris Beis
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Ioannis Kokkinopoulos
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece.
| |
Collapse
|
23
|
Ding Z, Tan K, Alter C, Temme S, Bouvain P, Owenier C, Hänsch S, Wesselborg S, Peter C, Weidtkamp-Peters S, Flögel U, Schira-Heinen J, Stühler K, Hesse J, Kögler G, Schrader J. Cardiac injection of USSC boosts remuscularization of the infarcted heart by shaping the T-cell response. J Mol Cell Cardiol 2023; 175:29-43. [PMID: 36493853 DOI: 10.1016/j.yjmcc.2022.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/20/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022]
Abstract
Regenerating the injured heart remains one of the most vexing challenges in cardiovascular medicine. Cell therapy has shown potential for treatment of myocardial infarction, but low cell retention so far has limited its success. Here we show that intramyocardial injection of highly apoptosis-resistant unrestricted somatic stem cells (USSC) into infarcted rat hearts resulted in an unprecedented thickening of the left ventricular wall with cTnT+/BrdU+ cardiomyocytes that was paralleled by progressively restored ejection fraction. USSC induced significant T-cell enrichment in ischemic tissue with enhanced expression of T-cell related cytokines. Inhibition of T-cell activation by anti-CD28 monoclonal antibody, fully abolished the regenerative response which was restored by adoptive T-cell transfer. Secretome analysis of USSC and lineage tracing studies suggest that USSC secrete paracrine factors over an extended period of time which boosts a T-cell driven endogenous regenerative response mainly from adult cardiomyocytes.
Collapse
Affiliation(s)
- Zhaoping Ding
- Department of Molecular Cardiology, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Kezhe Tan
- Department of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Christina Alter
- Department of Molecular Cardiology, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Sebastian Temme
- Department of Molecular Cardiology, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Pascal Bouvain
- Department of Molecular Cardiology, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Christoph Owenier
- Department of Molecular Cardiology, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Sebastian Hänsch
- Center for Advanced Imaging, Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | - Sebastian Wesselborg
- Institute of Molecular Medicine I, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Christoph Peter
- Institute of Molecular Medicine I, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | | | - Ulrich Flögel
- Department of Molecular Cardiology, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Jessica Schira-Heinen
- Molecular Proteomics Laboratory (MPL), Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory (MPL), Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Julia Hesse
- Department of Molecular Cardiology, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Gesine Kögler
- Jose Carreras Stem Cell Bank, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Jürgen Schrader
- Department of Molecular Cardiology, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany.
| |
Collapse
|
24
|
Fu X, Mishra R, Chen L, Arfat MY, Sharma S, Kingsbury T, Gunasekaran M, Saha P, Hong C, Yang P, Li D, Kaushal S. Exosomes mediated fibrogenesis in dilated cardiomyopathy through a MicroRNA pathway. iScience 2023; 26:105963. [PMID: 36818289 PMCID: PMC9932122 DOI: 10.1016/j.isci.2023.105963] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 11/02/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
Cardiac fibrosis is a hallmark in late-stage familial dilated cardiomyopathy (DCM) patients, although the underlying mechanism remains elusive. Cardiac exosomes (Exos) have been reported relating to fibrosis in ischemic cardiomyopathy. Thus, we investigated whether Exos secreted from the familial DCM cardiomyocytes could promote fibrogenesis. Using human iPSCs differentiated cardiomyocytes we isolated Exos of angiotensin II stimulation conditioned media from either DCM or control (CTL) cardiomyocytes. Of interest, cultured cardiac fibroblasts had increased fibrogenesis following exposure to DCM-Exos rather than CTL-Exos. Meanwhile, injecting DCM-Exos into mouse hearts enhanced cardiac fibrosis and impaired cardiac function. Mechanistically, we identified the upregulation of miRNA-218-5p in the DCM-Exos as a critical contributor to fibrogenesis. MiRNA-218-5p activated TGF-β signaling via suppression of TNFAIP3, a master inflammation inhibitor. In conclusion, our results illustrate a profibrotic effect of cardiomyocytes-derived Exos that highlights an additional pathogenesis pathway for cardiac fibrosis in DCM.
Collapse
Affiliation(s)
- Xuebin Fu
- Department of Cardiovascular-Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA,Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital, Chicago, IL, USA
| | - Rachana Mishra
- Department of Cardiovascular-Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA,Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital, Chicago, IL, USA
| | - Ling Chen
- Department of Cardiovascular-Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA,Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital, Chicago, IL, USA
| | - Mir Yasir Arfat
- Department of Cardiovascular-Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA,Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital, Chicago, IL, USA
| | - Sudhish Sharma
- Department of Cardiovascular-Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA,Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital, Chicago, IL, USA
| | - Tami Kingsbury
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Muthukumar Gunasekaran
- Department of Cardiovascular-Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA,Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital, Chicago, IL, USA
| | - Progyaparamita Saha
- Department of Cardiovascular-Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA,Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital, Chicago, IL, USA
| | - Charles Hong
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Peixin Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Deqiang Li
- Department of Surgery, Center for Vascular & Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA,Corresponding author
| | - Sunjay Kaushal
- Department of Cardiovascular-Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA,Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital, Chicago, IL, USA,Corresponding author
| |
Collapse
|
25
|
Wang T, Chen X, Wang K, Ju J, Yu X, Wang S, Liu C, Wang K. Cre-loxP-mediated genetic lineage tracing: Unraveling cell fate and origin in the developing heart. Front Cardiovasc Med 2023; 10:1085629. [PMID: 36923960 PMCID: PMC10008892 DOI: 10.3389/fcvm.2023.1085629] [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: 10/31/2022] [Accepted: 02/08/2023] [Indexed: 03/03/2023] Open
Abstract
The Cre-loxP-mediated genetic lineage tracing system is essential for constructing the fate mapping of single-cell progeny or cell populations. Understanding the structural hierarchy of cardiac progenitor cells facilitates unraveling cell fate and origin issues in cardiac development. Several prospective Cre-loxP-based lineage-tracing systems have been used to analyze precisely the fate determination and developmental characteristics of endocardial cells (ECs), epicardial cells, and cardiomyocytes. Therefore, emerging lineage-tracing techniques advance the study of cardiovascular-related cellular plasticity. In this review, we illustrate the principles and methods of the emerging Cre-loxP-based genetic lineage tracing technology for trajectory monitoring of distinct cell lineages in the heart. The comprehensive demonstration of the differentiation process of single-cell progeny using genetic lineage tracing technology has made outstanding contributions to cardiac development and homeostasis, providing new therapeutic strategies for tissue regeneration in congenital and cardiovascular diseases (CVDs).
Collapse
Affiliation(s)
- Tao Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Xinzhe Chen
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Kai Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Jie Ju
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Xue Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Shaocong Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Cuiyun Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Kun Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
26
|
Horowitz M, Hasin Y. Vascular compliance and left ventricular compliance cross talk: Implications for using long-term heat acclimation in cardiac care. Front Physiol 2023; 14:1074391. [PMID: 36960151 PMCID: PMC10027724 DOI: 10.3389/fphys.2023.1074391] [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: 10/19/2022] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
1) The first evidence of the beneficial impact of Long-Term-Heat-Acclimation (LTHA) on cardio-vascular compliance was the positive inotropic response and improved left ventricular (LV) compliance noted when isolated hearts from LTHA rats were studied. Human echo study demonstrates that passive HA affects the right ventricle and the atria as well. 2) There is a cross-talk between vascular and cardiac compliance. Vascular compliance per se is defined by central venous pressure-Blood volume relationship-Global Vascular Compliance (GVC). It is determined by the sum of the vascular compliance of the vessels in every organ in any physiological state, varies with LTHA and thus influences cardiac performance. LTHA improves endothelial function, increases NO (nitric oxide) production, in-turn stimulating alterations in ECM (extracellular matrix) via the TGF β1-SMAD pathway. 3) LTHA is associated with transformation from fast to slow myosin, heat acclimation ischemic/hypoxic cross-tolerance and alterations in the extracellular matrix. 4) A human translational study demonstrated improved LV compliance following bypass surgery in LTHA subjects compared to controls. 5) Diastolic dysfunction and the impact of comorbidities with vascular and non- vascular origins are major contributors to the syndrome of heart failure with preserved ejection function (HFPEF). Unfortunately, there is a paucity of treatment modalities that improve diastolic dysfunction. 6) In the current mini-review we suggest that LTHA may be beneficial to HFPEF patients by remodeling cardiac compliance and vascular response.
Collapse
Affiliation(s)
- Michal Horowitz
- Laboratory of Environmental Physiology, Faculty of Dentistry, The Hebrew University of Jerusalem, Jerusalem, Israel
- *Correspondence: Michal Horowitz,
| | | |
Collapse
|
27
|
Gui T, Burgering BMT. FOXOs: masters of the equilibrium. FEBS J 2022; 289:7918-7939. [PMID: 34610198 PMCID: PMC10078705 DOI: 10.1111/febs.16221] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/22/2021] [Accepted: 10/04/2021] [Indexed: 01/14/2023]
Abstract
Forkhead box O (FOXO) transcription factors (TFs) are a subclass of the larger family of forkhead TFs. Mammalians express four members FOXO1, FOXO3, FOXO4, and FOXO6. The interest in FOXO function stems mostly from their observed role in determining lifespan, where in model organisms, increased FOXO activity results in extended lifespan. FOXOs act as downstream of several signaling pathway and are extensively regulated through post-translational modifications. The transcriptional program activated by FOXOs in various cell types, organisms, and under various conditions has been described and has shed some light on what the critical transcriptional targets are in mediating FOXO function. At the cellular level, these studies have revealed a role for FOXOs in cell metabolism, cellular redox, cell proliferation, DNA repair, autophagy, and many more. The general picture that emerges hereof is that FOXOs act to preserve equilibrium, and they are important for cellular homeostasis. Here, we will first briefly summarize the general knowledge of FOXO regulation and possible functions. We will use genomic stability to illustrate how FOXOs ensure homeostasis. Genomic stability is critical for maintaining genetic integrity, and therefore preventing disease. However, genomic mutations need to occur during lifetime to enable evolution, yet their accumulation is believed to be causative to aging. Therefore, the role of FOXO in genomic stability may underlie its role in lifespan and aging. Finally, we will come up with questions on some of the unknowns in FOXO function, the answer(s) to which we believe will further our understanding of FOXO function and ultimately may help to understand lifespan and its consequences.
Collapse
Affiliation(s)
- Tianshu Gui
- Molecular Cancer Research, Center Molecular Medicine, University Medical Center Utrecht and the Oncode Institute, The Netherlands
| | - Boudewijn M T Burgering
- Molecular Cancer Research, Center Molecular Medicine, University Medical Center Utrecht and the Oncode Institute, The Netherlands
| |
Collapse
|
28
|
Nummi A, Pätilä T, Mulari S, Lampinen M, Nieminen T, Mäyränpää MI, Vento A, Harjula A, Kankuri E. Epicardial transplantation of autologous atrial appendage micrografts: evaluation of safety and feasibility in pigs after coronary artery occlusion. SCAND CARDIOVASC J 2022; 56:352-360. [PMID: 36002941 DOI: 10.1080/14017431.2022.2111462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/04/2022] [Accepted: 08/05/2022] [Indexed: 06/15/2023]
Abstract
Objectives. Several approaches devised for clinical utilization of cell-based therapies for heart failure often suffer from complex and lengthy preparation stages. Epicardial delivery of autologous atrial appendage micrografts (AAMs) with a clinically used extracellular matrix (ECM) patch provides a straightforward therapy alternative. We evaluated the operative feasibility and the effect of micrografts on the patch-induced epicardial foreign body inflammatory response in a porcine model of myocardial infarction. Design. Right atrial appendages were harvested and mechanically processed into AAMs. The left anterior descending coronary artery was ligated to generate acute infarction. Patches of ECM matrix with or without AAMs were transplanted epicardially onto the infarcted area. Four pigs received the ECM and four received the AAMs patch. Cardiac function was studied by echocardiography both preoperatively and at 3-week follow-up. The primary outcome measures were safety and feasibility of the therapy administration, and the secondary outcome was the inflammatory response to ECM. Results. Neither AAMs nor ECM patch-related complications were detected during the follow-up time. AAMs patch preparation was feasible according to time and safety. Inflammation was greatly reduced in AAMs when compared with ECM patches as measured by the amount of infiltrated inflammatory cells and area of inflammation. Immunohistochemistry demonstrated an increased CD3+ cell density in the AAMs patch infiltrate. Conclusions. Epicardial AAMs transplantation demonstrated safety and clinical feasibility. The use of micrografts significantly inhibited ECM-induced foreign body inflammatory reactivity. Transplantation of AAMs shows good clinical applicability as adjuvant therapy to cardiac surgery and can suppress acute inflammatory reactivity.
Collapse
Affiliation(s)
- Annu Nummi
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tommi Pätilä
- Pediatric Cardiac Surgery, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Severi Mulari
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Milla Lampinen
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tuomo Nieminen
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Päijät-Häme Joint Authority for Health and Wellbeing, Lahti, Finland
| | - Mikko I Mäyränpää
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Antti Vento
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Ari Harjula
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
29
|
Rojek K, Ćwiklińska M, Kuczak J, Guzowski J. Microfluidic Formulation of Topological Hydrogels for Microtissue Engineering. Chem Rev 2022; 122:16839-16909. [PMID: 36108106 PMCID: PMC9706502 DOI: 10.1021/acs.chemrev.1c00798] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Indexed: 02/07/2023]
Abstract
Microfluidics has recently emerged as a powerful tool in generation of submillimeter-sized cell aggregates capable of performing tissue-specific functions, so-called microtissues, for applications in drug testing, regenerative medicine, and cell therapies. In this work, we review the most recent advances in the field, with particular focus on the formulation of cell-encapsulating microgels of small "dimensionalities": "0D" (particles), "1D" (fibers), "2D" (sheets), etc., and with nontrivial internal topologies, typically consisting of multiple compartments loaded with different types of cells and/or biopolymers. Such structures, which we refer to as topological hydrogels or topological microgels (examples including core-shell or Janus microbeads and microfibers, hollow or porous microstructures, or granular hydrogels) can be precisely tailored with high reproducibility and throughput by using microfluidics and used to provide controlled "initial conditions" for cell proliferation and maturation into functional tissue-like microstructures. Microfluidic methods of formulation of topological biomaterials have enabled significant progress in engineering of miniature tissues and organs, such as pancreas, liver, muscle, bone, heart, neural tissue, or vasculature, as well as in fabrication of tailored microenvironments for stem-cell expansion and differentiation, or in cancer modeling, including generation of vascularized tumors for personalized drug testing. We review the available microfluidic fabrication methods by exploiting various cross-linking mechanisms and various routes toward compartmentalization and critically discuss the available tissue-specific applications. Finally, we list the remaining challenges such as simplification of the microfluidic workflow for its widespread use in biomedical research, bench-to-bedside transition including production upscaling, further in vivo validation, generation of more precise organ-like models, as well as incorporation of induced pluripotent stem cells as a step toward clinical applications.
Collapse
Affiliation(s)
- Katarzyna
O. Rojek
- Institute of Physical Chemistry, Polish Academy of Sciences, ul. Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Monika Ćwiklińska
- Institute of Physical Chemistry, Polish Academy of Sciences, ul. Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Julia Kuczak
- Institute of Physical Chemistry, Polish Academy of Sciences, ul. Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Jan Guzowski
- Institute of Physical Chemistry, Polish Academy of Sciences, ul. Kasprzaka 44/52, 01-224 Warsaw, Poland
| |
Collapse
|
30
|
Horak M, Fairweather D, Kokkonen P, Bednar D, Bienertova-Vasku J. Follistatin-like 1 and its paralogs in heart development and cardiovascular disease. Heart Fail Rev 2022; 27:2251-2265. [PMID: 35867287 PMCID: PMC11140762 DOI: 10.1007/s10741-022-10262-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2022] [Indexed: 11/29/2022]
Abstract
Cardiovascular diseases (CVDs) are a group of disorders affecting the heart and blood vessels and a leading cause of death worldwide. Thus, there is a need to identify new cardiokines that may protect the heart from damage as reported in GBD 2017 Causes of Death Collaborators (2018) (The Lancet 392:1736-1788). Follistatin-like 1 (FSTL1) is a cardiokine that is highly expressed in the heart and released to the serum after cardiac injury where it is associated with CVD and predicts poor outcome. The action of FSTL1 likely depends not only on the tissue source but also post-translation modifications that are target tissue- and cell-specific. Animal studies examining the effect of FSTL1 in various models of heart disease have exploded over the past 15 years and primarily report a protective effect spanning from inhibiting inflammation via transforming growth factor, preventing remodeling and fibrosis to promoting angiogenesis and hypertrophy. A better understanding of FSTL1 and its homologs is needed to determine whether this protein could be a useful novel biomarker to predict poor outcome and death and whether it has therapeutic potential. The aim of this review is to provide a comprehensive description of the literature for this family of proteins in order to better understand their role in normal physiology and CVD.
Collapse
Affiliation(s)
- Martin Horak
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
- Research Centre for Toxic Compounds in the Environment, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Piia Kokkonen
- Research Centre for Toxic Compounds in the Environment, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - David Bednar
- Research Centre for Toxic Compounds in the Environment, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Julie Bienertova-Vasku
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic.
- Research Centre for Toxic Compounds in the Environment, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic.
| |
Collapse
|
31
|
Peters MC, Maas RGC, van Adrichem I, Doevendans PAM, Mercola M, Šarić T, Buikema JW, van Mil A, Chamuleau SAJ, Sluijter JPG, Hnatiuk AP, Neef K. Metabolic Maturation Increases Susceptibility to Hypoxia-induced Damage in Human iPSC-derived Cardiomyocytes. Stem Cells Transl Med 2022; 11:1040-1051. [PMID: 36018047 PMCID: PMC9585948 DOI: 10.1093/stcltm/szac061] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 07/12/2022] [Indexed: 11/26/2022] Open
Abstract
The development of new cardioprotective approaches using in vivo models of ischemic heart disease remains challenging as differences in cardiac physiology, phenotype, and disease progression between humans and animals influence model validity and prognostic value. Furthermore, economical and ethical considerations have to be taken into account, especially when using large animal models with relevance for conducting preclinical studies. The development of human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) has opened new opportunities for in vitro studies on cardioprotective compounds. However, the immature cellular phenotype of iPSC-CMs remains a roadblock for disease modeling. Here, we show that metabolic maturation renders the susceptibility of iPSC-CMs to hypoxia further toward a clinically representative phenotype. iPSC-CMs cultured in a conventional medium did not show significant cell death after exposure to hypoxia. In contrast, metabolically matured (MM) iPSC-CMs showed inhibited mitochondrial respiration after exposure to hypoxia and increased cell death upon increased durations of hypoxia. Furthermore, we confirmed the applicability of MM iPSC-CMs for in vitro studies of hypoxic damage by validating the known cardioprotective effect of necroptosis inhibitor necrostatin-1. Our results provide important steps to improving and developing valid and predictive human in vitro models of ischemic heart disease.
Collapse
Affiliation(s)
- Marijn C Peters
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Renee G C Maas
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Iris van Adrichem
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Pieter A M Doevendans
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Mark Mercola
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA, USA
| | - Tomo Šarić
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jan W Buikema
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Alain van Mil
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Steven A J Chamuleau
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
- Department of Cardiology, Amsterdam UMC Heart Center, Amsterdam, The Netherlands
| | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Anna P Hnatiuk
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA, USA
| | - Klaus Neef
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
| |
Collapse
|
32
|
Kuwabara JT, Hara A, Bhutada S, Gojanovich GS, Chen J, Hokutan K, Shettigar V, Lee AY, DeAngelo LP, Heckl JR, Jahansooz JR, Tacdol DK, Ziolo MT, Apte SS, Tallquist MD. Consequences of PDGFRα + fibroblast reduction in adult murine hearts. eLife 2022; 11:e69854. [PMID: 36149056 PMCID: PMC9576271 DOI: 10.7554/elife.69854] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/22/2022] [Indexed: 12/01/2022] Open
Abstract
Fibroblasts produce the majority of collagen in the heart and are thought to regulate extracellular matrix (ECM) turnover. Although fibrosis accompanies many cardiac pathologies and is generally deleterious, the role of fibroblasts in maintaining the basal ECM network and in fibrosis in vivo is poorly understood. We genetically ablated fibroblasts in mice to evaluate the impact on homeostasis of adult ECM and cardiac function after injury. Fibroblast-ablated mice demonstrated a substantive reduction in cardiac fibroblasts, but fibrillar collagen and the ECM proteome were not overtly altered when evaluated by quantitative mass spectrometry and N-terminomics. However, the distribution and quantity of collagen VI, microfibrillar collagen that forms an open network with the basement membrane, was reduced. In fibroblast-ablated mice, cardiac function was better preserved following angiotensin II/phenylephrine (AngII/PE)-induced fibrosis and myocardial infarction (MI). Analysis of cardiomyocyte function demonstrated altered sarcomere shortening and slowed calcium decline in both uninjured and AngII/PE-infused fibroblast-ablated mice. After MI, the residual resident fibroblasts responded to injury, albeit with reduced proliferation and numbers immediately after injury. These results indicate that the adult mouse heart tolerates a significant degree of fibroblast loss with a potentially beneficial impact on cardiac function after injury. The cardioprotective effect of controlled fibroblast reduction may have therapeutic value in heart disease.
Collapse
Affiliation(s)
- Jill T Kuwabara
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Akitoshi Hara
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Sumit Bhutada
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research InstituteClevelandUnited States
| | - Greg S Gojanovich
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Jasmine Chen
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Kanani Hokutan
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Vikram Shettigar
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical CenterColumbusUnited States
| | - Anson Y Lee
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Lydia P DeAngelo
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Jack R Heckl
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Julia R Jahansooz
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Dillon K Tacdol
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| | - Mark T Ziolo
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical CenterColumbusUnited States
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research InstituteClevelandUnited States
| | - Michelle D Tallquist
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at ManoaHonoluluUnited States
| |
Collapse
|
33
|
Miller JM, Meki MH, Elnakib A, Ou Q, Abouleisa RRE, Tang XL, Salama ABM, Gebreil A, Lin C, Abdeltawab H, Khalifa F, Hill BG, Abi-Gerges N, Bolli R, El-Baz AS, Giridharan GA, Mohamed TMA. Biomimetic cardiac tissue culture model (CTCM) to emulate cardiac physiology and pathophysiology ex vivo. Commun Biol 2022; 5:934. [PMID: 36085302 PMCID: PMC9463130 DOI: 10.1038/s42003-022-03919-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
There is need for a reliable in vitro system that can accurately replicate the cardiac physiological environment for drug testing. The limited availability of human heart tissue culture systems has led to inaccurate interpretations of cardiac-related drug effects. Here, we developed a cardiac tissue culture model (CTCM) that can electro-mechanically stimulate heart slices with physiological stretches in systole and diastole during the cardiac cycle. After 12 days in culture, this approach partially improved the viability of heart slices but did not completely maintain their structural integrity. Therefore, following small molecule screening, we found that the incorporation of 100 nM tri-iodothyronine (T3) and 1 μM dexamethasone (Dex) into our culture media preserved the microscopic structure of the slices for 12 days. When combined with T3/Dex treatment, the CTCM system maintained the transcriptional profile, viability, metabolic activity, and structural integrity for 12 days at the same levels as the fresh heart tissue. Furthermore, overstretching the cardiac tissue induced cardiac hypertrophic signaling in culture, which provides a proof of concept for the ability of the CTCM to emulate cardiac stretch-induced hypertrophic conditions. In conclusion, CTCM can emulate cardiac physiology and pathophysiology in culture for an extended time, thereby enabling reliable drug screening.
Collapse
Affiliation(s)
- Jessica M Miller
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA
- Department of Bioengineering, University of Louisville, Louisville, USA
| | - Moustafa H Meki
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA
- Department of Bioengineering, University of Louisville, Louisville, USA
| | - Ahmed Elnakib
- Department of Bioengineering, University of Louisville, Louisville, USA
| | - Qinghui Ou
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA
| | - Riham R E Abouleisa
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA
| | - Xian-Liang Tang
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA
| | - Abou Bakr M Salama
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA
- Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ahmad Gebreil
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA
| | - Cindy Lin
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA
| | - Hisham Abdeltawab
- Department of Bioengineering, University of Louisville, Louisville, USA
| | - Fahmi Khalifa
- Department of Bioengineering, University of Louisville, Louisville, USA
| | - Bradford G Hill
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, USA
| | | | - Roberto Bolli
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA
| | - Ayman S El-Baz
- Department of Bioengineering, University of Louisville, Louisville, USA
| | | | - Tamer M A Mohamed
- From the Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, USA.
- Department of Bioengineering, University of Louisville, Louisville, USA.
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, USA.
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, USA.
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
34
|
Ali M, LaCanna R, Lian Z, Huang J, Tan Y, Shao W, Yu X, Tian Y. Transcriptional responses to injury of regenerative lung alveolar epithelium. iScience 2022; 25:104843. [PMID: 35996586 PMCID: PMC9391595 DOI: 10.1016/j.isci.2022.104843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/01/2022] [Accepted: 07/22/2022] [Indexed: 11/23/2022] Open
Abstract
The significance of alveolar epithelial type 2 (AT2) cell proliferation for lung alveolar epithelial homeostasis and regeneration after injury has been widely accepted. However, the heterogeneity of AT2 cell population for cell proliferation capacity remains disputed. By single-cell RNA sequencing and genetic lineage labeling using the Ki67 knock-in mouse model, we map all proliferative AT2 cells in homeostatic and regenerating murine lungs after injury induced by Streptococcus pneumoniae infection. The proliferative AT2 cell population displays a unique transcriptional program, which is regulated by activating transcription factor 3 (ATF3) and thyroid hormone receptor alpha (THRA) transcription factors. Overexpression of these two transcription factors in AT2 cells promoted AT2 cell proliferation and improved lung function after injury. These results indicate that increased expression of ATF3 and THRA at the onset of lung epithelial regeneration is required to permit rapid AT2 cell proliferation and hence progression through the recovery of lung epithelium.
Collapse
Affiliation(s)
- Mir Ali
- Department of Cardiovascular Sciences, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Ryan LaCanna
- Department of Cardiovascular Sciences, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Zhaorui Lian
- Coriell Institute for Medical Research, Camden, NJ 08103, USA
| | - Jian Huang
- Coriell Institute for Medical Research, Camden, NJ 08103, USA
| | - Yinfei Tan
- Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA 19111, USA
| | - Wenna Shao
- Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiang Yu
- Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ying Tian
- Department of Cardiovascular Sciences, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| |
Collapse
|
35
|
Cardiomyocyte-specific regression of nitrosative stress-mediated S-Nitrosylation of IKKγ alleviates pathological cardiac hypertrophy. Cell Signal 2022; 98:110403. [PMID: 35835332 DOI: 10.1016/j.cellsig.2022.110403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/21/2022] [Accepted: 07/07/2022] [Indexed: 11/21/2022]
Abstract
IKKγ prototypically promotes NFκBp65 activity by regulating the assembly of the IKK holocomplex. In hypertrophied cardiomyocytes, the p65-p300 complex-induced regenerative efforts are neutralized by the p53-p300 complex-mediated apoptotic load resulting in compromised cardiac function. The present study reports that nitrosative stress leads to S-Nitrosylation of IKKγ in hypertrophied cardiomyocytes in a pre-clinical model. Using a cardiomyocyte-targeted nanoconjugate, IKKγ S-Nitrosylation-resistant mutant plasmids were delivered to the pathologically hypertrophied heart that resulted in improved cardiac function by amelioration of cardiomyocyte apoptosis and simultaneous induction of their cell cycle re-entry machinery. Mechanistically, in IKKγ S-Nitrosyl mutant-transfected hypertrophied cells, increased IKKγ-p300 binding downregulated the binding of p53 and p65 with p300. This shifted the binding preference of p65 from p300 to HDAC1 resulting in upregulated expression of cyclin D1 and CDK2 via the p27/pRb pathway. This approach has therapeutic advantage over mainstream anti-hypertrophic remedies which concomitantly reduce the regenerative prowess of resident cardiomyocytes during hypertrophy upon downregulation of myocyte apoptosis. Therefore, cardiomyocyte-targeted delivery of IKKγ S-Nitrosyl mutants during hypertrophy can be exploited as a novel strategy to re-muscularize the diseased heart.
Collapse
|
36
|
Reboll MR, Klede S, Taft MH, Cai CL, Field LJ, Lavine KJ, Koenig AL, Fleischauer J, Meyer J, Schambach A, Niessen HW, Kosanke M, van den Heuvel J, Pich A, Bauersachs J, Wu X, Zheng L, Wang Y, Korf-Klingebiel M, Polten F, Wollert KC. Meteorin-like promotes heart repair through endothelial KIT receptor tyrosine kinase. Science 2022; 376:1343-1347. [PMID: 35709278 PMCID: PMC9838878 DOI: 10.1126/science.abn3027] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Effective tissue repair after myocardial infarction entails a vigorous angiogenic response, guided by incompletely defined immune cell-endothelial cell interactions. We identify the monocyte- and macrophage-derived cytokine METRNL (meteorin-like) as a driver of postinfarction angiogenesis and high-affinity ligand for the stem cell factor receptor KIT (KIT receptor tyrosine kinase). METRNL mediated angiogenic effects in cultured human endothelial cells through KIT-dependent signaling pathways. In a mouse model of myocardial infarction, METRNL promoted infarct repair by selectively expanding the KIT-expressing endothelial cell population in the infarct border zone. Metrnl-deficient mice failed to mount this KIT-dependent angiogenic response and developed severe postinfarction heart failure. Our data establish METRNL as a KIT receptor ligand in the context of ischemic tissue repair.
Collapse
Affiliation(s)
- Marc R. Reboll
- Division of Molecular and Translational Cardiology, Hans Borst Center for Heart and Stem Cell Research, Hannover Medical School; 30625 Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School; 30625 Hannover, Germany
| | - Stefanie Klede
- Division of Molecular and Translational Cardiology, Hans Borst Center for Heart and Stem Cell Research, Hannover Medical School; 30625 Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School; 30625 Hannover, Germany
| | - Manuel H. Taft
- Institute for Biophysical Chemistry, Hannover Medical School; 30625 Hannover, Germany
| | - Chen-Leng Cai
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine; Indianapolis, IN 46202, USA
| | - Loren J. Field
- Krannert Cardiovascular Research Center and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine; Indianapolis, IN 46202, USA
| | - Kory J. Lavine
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Andrew L. Koenig
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Jenni Fleischauer
- Institute of Experimental Hematology, Hannover Medical School; 30625 Hannover, Germany
| | - Johann Meyer
- Institute of Experimental Hematology, Hannover Medical School; 30625 Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School; 30625 Hannover, Germany
| | - Hans W. Niessen
- Department of Pathology and Department of Cardiac Surgery, Institute for Cardiovascular Research, University Medical Center; 1007 MB Amsterdam, The Netherlands
| | - Maike Kosanke
- Research Core Unit Genomics, Hannover Medical School; 30625 Hannover, Germany
| | - Joop van den Heuvel
- Technology Platform Recombinant Protein Expression, Helmholtz Center for Infection Research; 38124 Braunschweig, Germany
| | - Andreas Pich
- Core Unit Proteomics and Institute of Toxicology, Hannover Medical School; 30625 Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School; 30625 Hannover, Germany
| | - Xuekun Wu
- Division of Molecular and Translational Cardiology, Hans Borst Center for Heart and Stem Cell Research, Hannover Medical School; 30625 Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School; 30625 Hannover, Germany
| | - Linqun Zheng
- Division of Molecular and Translational Cardiology, Hans Borst Center for Heart and Stem Cell Research, Hannover Medical School; 30625 Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School; 30625 Hannover, Germany
| | - Yong Wang
- Division of Molecular and Translational Cardiology, Hans Borst Center for Heart and Stem Cell Research, Hannover Medical School; 30625 Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School; 30625 Hannover, Germany
| | - Mortimer Korf-Klingebiel
- Division of Molecular and Translational Cardiology, Hans Borst Center for Heart and Stem Cell Research, Hannover Medical School; 30625 Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School; 30625 Hannover, Germany
| | - Felix Polten
- Division of Molecular and Translational Cardiology, Hans Borst Center for Heart and Stem Cell Research, Hannover Medical School; 30625 Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School; 30625 Hannover, Germany
| | - Kai C. Wollert
- Division of Molecular and Translational Cardiology, Hans Borst Center for Heart and Stem Cell Research, Hannover Medical School; 30625 Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School; 30625 Hannover, Germany
| |
Collapse
|
37
|
Peters MC, Di Martino S, Boelens T, Qin J, van Mil A, Doevendans PA, Chamuleau SAJ, Sluijter JPG, Neef K. Follistatin-like 1 promotes proliferation of matured human hypoxic iPSC-cardiomyocytes and is secreted by cardiac fibroblasts. Mol Ther Methods Clin Dev 2022; 25:3-16. [PMID: 35317048 PMCID: PMC8917270 DOI: 10.1016/j.omtm.2022.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 02/19/2022] [Indexed: 12/12/2022]
Abstract
The human heart has limited regenerative capacity. Therefore, patients often progress to heart failure after ischemic injury, despite advances in reperfusion therapies generally decreasing mortality. Depending on its glycosylation state, Follistatin-like 1 (FSTL1) has been shown to increase cardiomyocyte (CM) proliferation, decrease CM apoptosis, and prevent cardiac rupture in animal models of ischemic heart disease. To explore its therapeutic potential, we used a human in vitro model of cardiac ischemic injury with human induced pluripotent stem cell-derived CMs (iPSC-CMs) and assessed regenerative effects of two differently glycosylated variants of human FSTL1. Furthermore, we investigated the FSTL1-mediated interplay between human cardiac fibroblasts (cFBs) and iPSC-CMs in hypoxia. Both FSTL1 variants increased viability, while only hypo-glycosylated FSTL1 increased CM proliferation post-hypoxia. Human fetal cardiac fibroblasts (fcFBs) expressed and secreted FSTL1 under normoxic conditions, while FSTL1 secretion increased by iPSC-cFBs upon hypoxia but decreased in iPSC-CMs. Co-culture of iPSC-CMs and cFBs increased FSTL1 secretion compared with cFB mono-culture. Taken together, we confirm that FSTL1 induces iPSC-CM proliferation in a human cardiac in vitro hypoxia damage model. Furthermore, we show hypoxia-related FSTL1 secretion by human cFBs and indications for FSTL1-mediated intercellular communication between cardiac cell types in response to hypoxic conditions.
Collapse
Affiliation(s)
- Marijn C Peters
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Sofia Di Martino
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Thomas Boelens
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Jiabin Qin
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Alain van Mil
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Pieter A Doevendans
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Steven A J Chamuleau
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands.,Department of Cardiology, Amsterdam Medical Centre, 1105 AZ Amsterdam, the Netherlands
| | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Klaus Neef
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| |
Collapse
|
38
|
Desmin deficiency affects the microenvironment of the cardiac side population and Sca1+ stem cell population of the adult heart and impairs their cardiomyogenic commitment. Cell Tissue Res 2022; 389:309-326. [DOI: 10.1007/s00441-022-03643-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/20/2022] [Indexed: 11/02/2022]
|
39
|
Wehrens M, de Leeuw AE, Wright-Clark M, Eding JEC, Boogerd CJ, Molenaar B, van der Kraak PH, Kuster DWD, van der Velden J, Michels M, Vink A, van Rooij E. Single-cell transcriptomics provides insights into hypertrophic cardiomyopathy. Cell Rep 2022; 39:110809. [PMID: 35545053 DOI: 10.1016/j.celrep.2022.110809] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/25/2022] [Accepted: 04/21/2022] [Indexed: 11/24/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a genetic heart disease that is characterized by unexplained segmental hypertrophy that is usually most pronounced in the septum. While sarcomeric gene mutations are often the genetic basis for HCM, the mechanistic origin for the heterogeneous remodeling remains largely unknown. A better understanding of the gene networks driving the cardiomyocyte (CM) hypertrophy is required to improve therapeutic strategies. Patients suffering from HCM often receive a septal myectomy surgery to relieve outflow tract obstruction due to hypertrophy. Using single-cell RNA sequencing (scRNA-seq) on septal myectomy samples from patients with HCM, we identify functional links between genes, transcription factors, and cell size relevant for HCM. The data show the utility of using scRNA-seq on the human hypertrophic heart, highlight CM heterogeneity, and provide a wealth of insights into molecular events involved in HCM that can eventually contribute to the development of enhanced therapies.
Collapse
Affiliation(s)
- Martijn Wehrens
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center, Utrecht, the Netherlands
| | - Anne E de Leeuw
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center, Utrecht, the Netherlands
| | - Maya Wright-Clark
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center, Utrecht, the Netherlands; Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Joep E C Eding
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center, Utrecht, the Netherlands
| | - Cornelis J Boogerd
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center, Utrecht, the Netherlands
| | - Bas Molenaar
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center, Utrecht, the Netherlands
| | - Petra H van der Kraak
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Diederik W D Kuster
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Michelle Michels
- Department of Cardiology, Erasmus MC, Rotterdam, the Netherlands
| | - Aryan Vink
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center, Utrecht, the Netherlands; Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
40
|
Prasad V, Molkentin JD. At the Brink of Human Therapy to Generate New Myocytes in the Adult Injured Heart. Circulation 2022; 145:1356-1358. [PMID: 35467953 PMCID: PMC9046981 DOI: 10.1161/circulationaha.122.059106] [Citation(s) in RCA: 0] [Impact Index Per Article: 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)
- Vikram Prasad
- The University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Jeffery D. Molkentin
- The University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
41
|
Anto Michel N, Ljubojevic-Holzer S, Bugger H, Zirlik A. Cellular Heterogeneity of the Heart. Front Cardiovasc Med 2022; 9:868466. [PMID: 35548426 PMCID: PMC9081371 DOI: 10.3389/fcvm.2022.868466] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/23/2022] [Indexed: 11/18/2022] Open
Abstract
Recent advances in technology such as the introduction of high throughput multidimensional tools like single cell sequencing help to characterize the cellular composition of the human heart. The diversity of cell types that has been uncovered by such approaches is by far greater than ever expected before. Accurate identification of the cellular variety and dynamics will not only facilitate a much deeper understanding of cardiac physiology but also provide important insights into mechanisms underlying its pathological transformation. Distinct cellular patterns of cardiac cell clusters may allow differentiation between a healthy heart and a sick heart while potentially predicting future disease at much earlier stages than currently possible. These advances have already extensively improved and will ultimately revolutionize our knowledge of the mechanisms underlying cardiovascular disease as such. In this review, we will provide an overview of the cells present in the human and rodent heart as well as genes that may be used for their identification.
Collapse
|
42
|
Sano T, Ito T, Ishigami S, Bandaru S, Sano S. Intrinsic activation of cardiosphere-derived cells enhances myocardial repair. J Thorac Cardiovasc Surg 2022; 163:1479-1490.e5. [PMID: 32682583 DOI: 10.1016/j.jtcvs.2020.05.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 05/01/2020] [Accepted: 05/01/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Permanent loss of cardiomyocytes after myocardial infarction results in irreversible damage to cardiac function. The present study aims to enhance the cardiomyogenic efficiency of cardiosphere-derived cells (CDCs) to develop into large populations of cardiomyocytes by intrinsic activation of cardio-specific differentiation factors (Gata4, Mef2c, Nkx2-5, Hand2, and Tnnt2) by a CRISPR/dCas9 assisted transcriptional enhancement system. METHODS Exhaustive screening was performed to identify the specific sequences in endogenous regulatory regions (enhancers and promoters) responsible for transcriptional activation of the target genes, via a CRISPR/dCas9 system fused with transcriptional activator VP64 (CRISPR-dCas9-VP64). In a rat model of acute myocardial infarction, we compared the regenerative potential and functional benefits of CDCs with or without transcriptional activation. RESULTS We identified a panel of specific CRISPR RNA targeting the enhancers and promoters, which demonstrated significantly higher expression of differentiation factors of Gata4, Hand2, and Tnnt2. The group of CDCs with transcriptional activator VP64 (CDC with VP64) showed significant improvement in the left ventricular ejection fraction (61.9% vs 52.5% and 44.1% in the CDC without transcriptional activation group and control) and decreased scar area in the heart. CONCLUSIONS We have identified endogenous regulatory regions responsible for an intrinsic activation of cardio-specific differentiation factors assisted via a CRISPR/dCas9 gene transcriptional system. The CRISPR/dCas9 system may provide an efficient and effective means of regulating Tnnt2 gene activation within stem cells. Subsequently, this system can be used to enhance transplanted CDCs differentiation potential within ischemic myocardia to better therapeutic outcomes of patients with ischemic heart disease.
Collapse
Affiliation(s)
- Toshikazu Sano
- Division of Pediatric Cardiothoracic Surgery, Department of Surgery, University of California at San Francisco, San Francisco, Calif
| | - Tatsuo Ito
- Department of Hygiene, Kawasaki Medical University, Okayama, Japan
| | - Shuta Ishigami
- Division of Pediatric Cardiothoracic Surgery, Department of Surgery, University of California at San Francisco, San Francisco, Calif
| | - Srinivas Bandaru
- Department of Hygiene, Kawasaki Medical University, Okayama, Japan
| | - Shunji Sano
- Division of Pediatric Cardiothoracic Surgery, Department of Surgery, University of California at San Francisco, San Francisco, Calif.
| |
Collapse
|
43
|
Regulation of Epicardial Cell Fate during Cardiac Development and Disease: An Overview. Int J Mol Sci 2022; 23:ijms23063220. [PMID: 35328640 PMCID: PMC8950551 DOI: 10.3390/ijms23063220] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 01/27/2023] Open
Abstract
The epicardium is the outermost cell layer in the vertebrate heart that originates during development from mesothelial precursors located in the proepicardium and septum transversum. The epicardial layer plays a key role during cardiogenesis since a subset of epicardial-derived cells (EPDCs) undergo an epithelial–mesenchymal transition (EMT); migrate into the myocardium; and differentiate into distinct cell types, such as coronary vascular smooth muscle cells, cardiac fibroblasts, endothelial cells, and presumably a subpopulation of cardiomyocytes, thus contributing to complete heart formation. Furthermore, the epicardium is a source of paracrine factors that support cardiac growth at the last stages of cardiogenesis. Although several lineage trace studies have provided some evidence about epicardial cell fate determination, the molecular mechanisms underlying epicardial cell heterogeneity remain not fully understood. Interestingly, seminal works during the last decade have pointed out that the adult epicardium is reactivated after heart damage, re-expressing some embryonic genes and contributing to cardiac remodeling. Therefore, the epicardium has been proposed as a potential target in the treatment of cardiovascular disease. In this review, we summarize the previous knowledge regarding the regulation of epicardial cell contribution during development and the control of epicardial reactivation in cardiac repair after damage.
Collapse
|
44
|
Cedervall J, Herre M, Dragomir A, Rabelo-Melo F, Svensson A, Thålin C, Rosell A, Hjalmar V, Wallén H, Lindman H, Pejler G, Hagström E, Hultström M, Larsson A, Olsson AK. Neutrophil extracellular traps promote cancer-associated inflammation and myocardial stress. Oncoimmunology 2022; 11:2049487. [PMID: 35309730 PMCID: PMC8928831 DOI: 10.1080/2162402x.2022.2049487] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cancer is associated with systemic pathologies that contribute to mortality, such as thrombosis and distant organ failure. The aim of this study was to investigate the potential role of neutrophil extracellular traps (NETs) in myocardial inflammation and tissue damage in treatment-naïve individuals with cancer. Mice with mammary carcinoma (MMTV-PyMT) had increased plasma levels of NETs measured as H3Cit-DNA complexes, paralleled with elevated coagulation, compared to healthy littermates. MMTV-PyMT mice displayed upregulation of pro-inflammatory markers in the heart, myocardial hypertrophy and elevated cardiac disease biomarkers in the blood, but not echocardiographic heart failure. Moreover, increased endothelial proliferation was observed in hearts from tumor-bearing mice. Removal of NETs by DNase I treatment suppressed the myocardial inflammation, expression of cardiac disease biomarkers and endothelial proliferation. Compared to a healthy control group, treatment-naïve cancer patients with different malignant disorders had increased NET formation, which correlated to plasma levels of the inflammatory marker CRP and the cardiac disease biomarkers NT-proBNP and sTNFR1, in agreement with the mouse data. Altogether, our data indicate that NETs contribute to inflammation and myocardial stress during malignancy. These findings suggest NETs as potential therapeutic targets to prevent cardiac inflammation and dysfunction in cancer patients.
Collapse
Affiliation(s)
- J. Cedervall
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden
| | - M. Herre
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden
| | - A. Dragomir
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - F. Rabelo-Melo
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden
| | - A. Svensson
- Department of Medical Cell Biology, Integrative Physiology, Uppsala University, Uppsala, Sweden
- Department of Surgical Sciences, Anaesthesiology and Intensive Care Medicine, Uppsala University, Uppsala, Sweden
| | - C. Thålin
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - A. Rosell
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - V. Hjalmar
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
- Diagnostic Centre, Danderyd Hospital, Karolinska Institute, Stockholm, Sweden
| | - H. Wallén
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - H. Lindman
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - G. Pejler
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden
| | - E. Hagström
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - M. Hultström
- Department of Medical Cell Biology, Integrative Physiology, Uppsala University, Uppsala, Sweden
- Department of Surgical Sciences, Anaesthesiology and Intensive Care Medicine, Uppsala University, Uppsala, Sweden
| | - A. Larsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - AK. Olsson
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden
| |
Collapse
|
45
|
Tu SM, Pisters LL. Stem-Cell Theory of Cancer: Implications for Antiaging and Anticancer Strategies. Cancers (Basel) 2022; 14:1338. [PMID: 35267646 PMCID: PMC8909197 DOI: 10.3390/cancers14051338] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/16/2022] [Accepted: 02/28/2022] [Indexed: 12/03/2022] Open
Abstract
A stem-cell theory of cancer predicates that not only does the cell affect the niche, the niche also affects the cell. It implicates that even though genetic makeup may be supreme, cellular context is key. When we attempt to solve the mystery of a long cancer-free life, perhaps we need to search no further than the genetics and epigenetics of the naked mole-rat. When we try to unlock the secrets in the longevity and quality of life, perhaps we need to look no further than the lifestyle and habits of the super centenarians. We speculate that people with Down's syndrome and progeria age faster but have fewer cancers, because they are depleted of stem cells, and, as a consequence, have fewer opportunities for stem cell defects that could predispose them to the development of cancer. We contemplate whether these incredible experiments of nature may provide irrefutable evidence that cancer is a stem-cell disease-fewer aberrant stem cells, fewer cancers; no defective stem cells, no cancer. In this perspective, we investigate a stem-cell origin of aging and cancer. We elaborate an intriguing inverse relationship between longevity and malignancy in the naked mole-rat, in Down's syndrome, and in progeria. We postulate that stem-cell pools and stemness factors may affect aging and dictate cancer. We propose that a healthy microbiome may protect and preserve stem cell reserves and provide meaningful antiaging effects and anticancer benefits.
Collapse
Affiliation(s)
- Shi-Ming Tu
- Division of Hematology/Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Louis L. Pisters
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| |
Collapse
|
46
|
In Vivo Methods to Monitor Cardiomyocyte Proliferation. J Cardiovasc Dev Dis 2022; 9:jcdd9030073. [PMID: 35323621 PMCID: PMC8950582 DOI: 10.3390/jcdd9030073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 12/07/2022] Open
Abstract
Adult mammalian cardiomyocytes demonstrate scarce cycling and even lower proliferation rates in response to injury. Signals that enhance cardiomyocyte proliferation after injury will be groundbreaking, address unmet clinical needs, and represent new strategies to treat cardiovascular diseases. In vivo methods to monitor cardiomyocyte proliferation are critical to addressing this challenge. Fortunately, advances in transgenic approaches provide sophisticated techniques to quantify cardiomyocyte cycling and proliferation.
Collapse
|
47
|
Arslan U, Moruzzi A, Nowacka J, Mummery CL, Eckardt D, Loskill P, Orlova VV. Microphysiological stem cell models of the human heart. Mater Today Bio 2022; 14:100259. [PMID: 35514437 PMCID: PMC9062349 DOI: 10.1016/j.mtbio.2022.100259] [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: 03/03/2022] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 11/10/2022] Open
Abstract
Models of heart disease and drug responses are increasingly based on human pluripotent stem cells (hPSCs) since their ability to capture human heart (dys-)function is often better than animal models. Simple monolayer cultures of hPSC-derived cardiomyocytes, however, have shortcomings. Some of these can be overcome using more complex, multi cell-type models in 3D. Here we review modalities that address this, describe efforts to tailor readouts and sensors for monitoring tissue- and cell physiology (exogenously and in situ) and discuss perspectives for implementation in industry and academia.
Collapse
Affiliation(s)
- Ulgu Arslan
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Alessia Moruzzi
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- Institute for Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Joanna Nowacka
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, the Netherlands
| | | | - Peter Loskill
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- Institute for Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
- 3R-Center for in Vitro Models and Alternatives to Animal Testing, Tübingen, Germany
| | - Valeria V. Orlova
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, the Netherlands
| |
Collapse
|
48
|
Zhou Y, Ng DY, Richards AM, Wang P. Loss of full-length pumilio 1 abrogates miRNA-221-induced gene p27 silencing-mediated cell proliferation in the heart. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 27:456-470. [PMID: 35036057 PMCID: PMC8728526 DOI: 10.1016/j.omtn.2021.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 12/09/2021] [Indexed: 12/02/2022]
Abstract
Upregulated expression of microRNA (miR)-221 is associated with downregulation of p27 and subsequent increased cell proliferation in a variety of human cancers. It is unknown whether miR-221 mimics could trigger neoplastic cellular proliferation. In vitro, we demonstrated miR-221 significantly downregulates the expression of P27 and increases proliferation of H9c2 and cardiac fibroblasts. The knockdown of PUM1 but not PUM2 abolished such effects by miR-221, as verified by RT-qPCR and western blot, direct binding of p27 3′ UTR by luciferase reporter assay and cell proliferation by Ki67. In vivo expression of P27 in the rat liver, heart, kidney, spleen, and muscle were not affected by miR-221 at 1 and 4 mg/kg and concurrently full-length (FL) PUM1 (140 kDa) was not detected. Instead, isoforms of 105 and 90 kDa were observed and generated through alternative RNA slicing verified by cDNA cloning and sequencing and cathepsin K cleavage confirmed by studies with the inhibitor odanacatib. This is the first study to address the possible pro-proliferative effects of miR-221 mimic therapeutics in cardiovascular applications. Loss of FL PUM1 expression is a key factor abrogating miR-221-mediated p27 regulation, although other concurrent mechanisms cannot be excluded. Our findings provide essential insights into the context-dependent nature of miRNA functionality.
Collapse
|
49
|
Tian X, Zhou B. Coronary vessel formation in development and regeneration: origins and mechanisms. J Mol Cell Cardiol 2022; 167:67-82. [DOI: 10.1016/j.yjmcc.2022.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 03/12/2022] [Accepted: 03/22/2022] [Indexed: 10/18/2022]
|
50
|
Herrera-Rivero M, Gandhi S, Witten A, Ghalawinji A, Schotten U, Stoll M. Cardiac chamber-specific genetic alterations suggest candidate genes and pathways implicating the left ventricle in the pathogenesis of atrial fibrillation. Genomics 2022; 114:110320. [PMID: 35218871 DOI: 10.1016/j.ygeno.2022.110320] [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: 10/19/2021] [Revised: 01/12/2022] [Accepted: 02/19/2022] [Indexed: 11/15/2022]
Abstract
It is believed that the atria play a predominant role in the initiation and maintenance of atrial fibrillation (AF), while the role of left ventricular dysfunction in the pathophysiology remains enigmatic. We sought to dissect chamber specificity of AF-associated transcriptional changes using RNA-sequencing. We performed intra- and inter-chamber differential expression analyses comparing AF against sinus rhythm to identify genes specifically dysregulated in human left atria, right atria, and left ventricle (LV), and integrated known AF genetic associations with expression quantitative trait loci datasets to inform the potential for disease causal contributions within each chamber. Inter-chamber patterns changed drastically. Vast AF-associated transcriptional changes specific to LV, enriched for biological pathway terms implicating mitochondrial function, developmental processes and immunity, were supported at the genetic level, but no major enrichments for candidate genes specific to the atria were found. Our observations suggest an active role of the LV in the pathogenesis of AF.
Collapse
Affiliation(s)
- Marisol Herrera-Rivero
- Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany
| | - Shrey Gandhi
- Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany; Institute for Evolution and Biodiversity, University of Münster, Münster, Germany
| | - Anika Witten
- Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany
| | - Amer Ghalawinji
- Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany
| | - Ulrich Schotten
- Department of Physiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Monika Stoll
- Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany; Department of Biochemistry, Genetic Epidemiology and Statistical Genetics, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|