1
|
Yuan J, Deng T, Yang Q, Lv D, Zhou Z, You L, Feng Q, Meng X, Pang Q, Li H, Zhu B. Loss of LSD1 ameliorates myocardial infarction by regulating angiogenesis via transcriptional activation of Vegfa. Life Sci 2025; 372:123613. [PMID: 40210117 DOI: 10.1016/j.lfs.2025.123613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/13/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025]
Abstract
AIMS Our study aims to explore the regulatory role and underlying mechanisms of Lysine-specific demethylase 1 (LSD1) in angiogenesis following myocardial infarction (MI). MATERIALS AND METHODS We generated inducible cardiomyocyte-specific Lsd1 knockout (Lsd1-cKO) mice and established a MI model. The function of LSD1 in cardiac angiogenesis in MI mice was assessed through echocardiography, histopathological staining, and immunofluorescence analysis. In vitro, Lsd1 silencing in cardiomyocytes was achieved by transfecting small interfering RNA (siRNA), followed by hypoxic treatment to simulate the in vivo MI model. The above cardiomyocyte-conditioned medium was collected and used to treat endothelial cells to observe changes in endothelial function. Additionally, we employed Cleavage Under Targets and Tagmentation sequencing (CUT&Tag-seq) to investigate the potential mechanisms by which LSD1 exerts its effects. KEY FINDINGS We found that the absence of LSD1 protected against cardiac dysfunction and promoted angiogenesis in mice with MI. Lsd1-silenced cardiomyocytes enhance the migration and tube formation function of endothelial cells by releasing vascular endothelial growth factor A (VEGF-A) under hypoxic conditions. The combined analysis of CUT&Tag-seq data revealed that silencing of Lsd1 promoted the monomethylation of H3K4 at the Vegfa promoter and region, leading to the transcriptional activation of Vegfa mRNA in cardiomyocytes. SIGNIFICANCE Our research indicates that lowered level of LSD1 in cardiomyocytes enhances VEGF-A paracrine secretion and improves endothelial cell function through cross-talk, ultimately promoting angiogenesis. These findings suggest that targeting LSD1 might be an effective therapeutic approach to protect against MI.
Collapse
Affiliation(s)
- Jinghan Yuan
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tian Deng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qingshan Yang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Danyi Lv
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhenfang Zhou
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lu You
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qipu Feng
- Animal Experiment Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Xiangmin Meng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiuyu Pang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hao Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bingmei Zhu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
2
|
Liberale L, Tual-Chalot S, Sedej S, Ministrini S, Georgiopoulos G, Grunewald M, Bäck M, Bochaton-Piallat ML, Boon RA, Ramos GC, de Winther MPJ, Drosatos K, Evans PC, Ferguson JF, Forslund-Startceva SK, Goettsch C, Giacca M, Haendeler J, Kallikourdis M, Ketelhuth DFJ, Koenen RR, Lacolley P, Lutgens E, Maffia P, Miwa S, Monaco C, Montecucco F, Norata GD, Osto E, Richardson GD, Riksen NP, Soehnlein O, Spyridopoulos I, Van Linthout S, Vilahur G, Wentzel JJ, Andrés V, Badimon L, Benetos A, Binder CJ, Brandes RP, Crea F, Furman D, Gorbunova V, Guzik TJ, Hill JA, Lüscher TF, Mittelbrunn M, Nencioni A, Netea MG, Passos JF, Stamatelopoulos KS, Tavernarakis N, Ungvari Z, Wu JC, Kirkland JL, Camici GG, Dimmeler S, Kroemer G, Abdellatif M, Stellos K. Roadmap for alleviating the manifestations of ageing in the cardiovascular system. Nat Rev Cardiol 2025:10.1038/s41569-025-01130-5. [PMID: 39972009 DOI: 10.1038/s41569-025-01130-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2025] [Indexed: 02/21/2025]
Abstract
Ageing of the cardiovascular system is associated with frailty and various life-threatening diseases. As global populations grow older, age-related conditions increasingly determine healthspan and lifespan. The circulatory system not only supplies nutrients and oxygen to all tissues of the human body and removes by-products but also builds the largest interorgan communication network, thereby serving as a gatekeeper for healthy ageing. Therefore, elucidating organ-specific and cell-specific ageing mechanisms that compromise circulatory system functions could have the potential to prevent or ameliorate age-related cardiovascular diseases. In support of this concept, emerging evidence suggests that targeting the circulatory system might restore organ function. In this Roadmap, we delve into the organ-specific and cell-specific mechanisms that underlie ageing-related changes in the cardiovascular system. We raise unanswered questions regarding the optimal design of clinical trials, in which markers of biological ageing in humans could be assessed. We provide guidance for the development of gerotherapeutics, which will rely on the technological progress of the diagnostic toolbox to measure residual risk in elderly individuals. A major challenge in the quest to discover interventions that delay age-related conditions in humans is to identify molecular switches that can delay the onset of ageing changes. To overcome this roadblock, future clinical trials need to provide evidence that gerotherapeutics directly affect one or several hallmarks of ageing in such a manner as to delay, prevent, alleviate or treat age-associated dysfunction and diseases.
Collapse
Affiliation(s)
- Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK.
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | | | - Myriam Grunewald
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Magnus Bäck
- Translational Cardiology, Centre for Molecular Medicine, Department of Medicine Solna, and Department of Cardiology, Heart and Vascular Centre, Karolinska Institutet, Stockholm, Sweden
- Inserm, DCAC, Université de Lorraine, Nancy, France
| | | | - Reinier A Boon
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC location VUmc, Amsterdam, Netherlands
| | - Gustavo Campos Ramos
- Department of Internal Medicine I/Comprehensive Heart Failure Centre, University Hospital Würzburg, Würzburg, Germany
| | - Menno P J de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences: Atherosclerosis and Ischaemic Syndromes; Amsterdam Infection and Immunity: Inflammatory Diseases, Amsterdam UMC location AMC, Amsterdam, Netherlands
| | - Konstantinos Drosatos
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Paul C Evans
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jane F Ferguson
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sofia K Forslund-Startceva
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Goettsch
- Department of Internal Medicine I, Division of Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Mauro Giacca
- British Heart foundation Centre of Reseach Excellence, King's College London, London, UK
| | - Judith Haendeler
- Cardiovascular Degeneration, Medical Faculty, University Hospital and Heinrich-Heine University, Düsseldorf, Germany
| | - Marinos Kallikourdis
- Adaptive Immunity Lab, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Daniel F J Ketelhuth
- Cardiovascular and Renal Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Rory R Koenen
- CARIM-School for Cardiovascular Diseases, Department of Biochemistry, Maastricht University, Maastricht, Netherlands
| | | | - Esther Lutgens
- Department of Cardiovascular Medicine & Immunology, Mayo Clinic, Rochester, MN, USA
| | - Pasquale Maffia
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Satomi Miwa
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Claudia Monaco
- Kennedy Institute, NDORMS, University of Oxford, Oxford, UK
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Osto
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Gavin D Richardson
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Oliver Soehnlein
- Institute of Experimental Pathology, University of Münster, Münster, Germany
| | - Ioakim Spyridopoulos
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Sophie Van Linthout
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätmedizin Berlin, Berlin, Germany
| | - Gemma Vilahur
- Research Institute, Hospital de la Santa Creu y Sant Pau l, IIB-Sant Pau, Barcelona, Spain
| | - Jolanda J Wentzel
- Cardiology, Biomedical Engineering, Erasmus MC, Rotterdam, Netherlands
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), CIBERCV, Madrid, Spain
| | - Lina Badimon
- Cardiovascular Health and Innovation Research Foundation (FICSI) and Cardiovascular Health and Network Medicine Department, University of Vic (UVIC-UCC), Barcelona, Spain
| | - Athanase Benetos
- Department of Geriatrics, University Hospital of Nancy and Inserm DCAC, Université de Lorraine, Nancy, France
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany
| | - Filippo Crea
- Centre of Excellence of Cardiovascular Sciences, Ospedale Isola Tiberina - Gemelli Isola, Roma, Italy
| | - David Furman
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK
| | - Joseph A Hill
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Thomas F Lüscher
- Heart Division, Royal Brompton and Harefield Hospital and National Heart and Lung Institute, Imperial College, London, UK
| | - María Mittelbrunn
- Consejo Superior de Investigaciones Científicas (CSIC), Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Alessio Nencioni
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- Dipartimento di Medicina Interna e Specialità Mediche-DIMI, Università degli Studi di Genova, Genova, Italy
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - João F Passos
- Department of Physiology and Biomedical Engineering, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Kimon S Stamatelopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nektarios Tavernarakis
- Medical School, University of Crete, and Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Zoltan Ungvari
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - James L Kirkland
- Center for Advanced Gerotherapeutics, Division of Endocrinology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm, Institut Universitaire de France, Paris, France
| | | | - Konstantinos Stellos
- Department of Cardiovascular Research, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
3
|
Panico C, Felicetta A, Kunderfranco P, Cremonesi M, Salvarani N, Carullo P, Colombo F, Idini A, Passaretti M, Doro R, Rubino M, Villaschi A, Da Rin G, Peano C, Kallikourdis M, Greco CM, Condorelli G. Single-Cell RNA Sequencing Reveals Metabolic Stress-Dependent Activation of Cardiac Macrophages in a Model of Dyslipidemia-Induced Diastolic Dysfunction. Circulation 2024; 150:1517-1532. [PMID: 38126199 DOI: 10.1161/circulationaha.122.062984] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/17/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Metabolic distress is often associated with heart failure with preserved ejection fraction (HFpEF) and represents a therapeutic challenge. Metabolism-induced systemic inflammation links comorbidities with HFpEF. How metabolic changes affect myocardial inflammation in the context of HFpEF is not known. METHODS We found that ApoE knockout mice fed a Western diet recapitulate many features of HFpEF. Single-cell RNA sequencing was used for expression analysis of CD45+ cardiac cells to evaluate the involvement of inflammation in diastolic dysfunction. We focused bioinformatics analysis on macrophages, obtaining high-resolution identification of subsets of these cells in the heart, enabling us to study the outcomes of metabolic distress on the cardiac macrophage infiltrate and to identify a macrophage-to-cardiomyocyte regulatory axis. To test whether a clinically relevant sodium glucose cotransporter-2 inhibitor could ameliorate the cardiac immune infiltrate profile in our model, mice were randomized to receive the sodium glucose cotransporter-2 inhibitor dapagliflozin or vehicle for 8 weeks. RESULTS ApoE knockout mice fed a Western diet presented with reduced diastolic function, reduced exercise tolerance, and increased pulmonary congestion associated with cardiac lipid overload and reduced polyunsaturated fatty acids. The main immune cell types infiltrating the heart included 4 subpopulations of resident and monocyte-derived macrophages, determining a proinflammatory profile exclusively in ApoE knockout-Western diet mice. Lipid overload had a direct effect on inflammatory gene activation in macrophages, mediated through endoplasmic reticulum stress pathways. Investigation of the macrophage-to-cardiomyocyte regulatory axis revealed the potential effects on cardiomyocytes of multiple inflammatory cytokines secreted by macrophages, affecting pathways such as hypertrophy, fibrosis, and autophagy. Finally, we describe an anti-inflammatory effect of sodium glucose cotransporter-2 inhibition in this model. CONCLUSIONS Using single-cell RNA sequencing in a model of diastolic dysfunction driven by hyperlipidemia, we have determined the effects of metabolic distress on cardiac inflammatory cells, in particular on macrophages, and suggest sodium glucose cotransporter-2 inhibitors as potential therapeutic agents for the targeting of a specific phenotype of HFpEF.
Collapse
Affiliation(s)
- Cristina Panico
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Arianna Felicetta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Paolo Kunderfranco
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Marco Cremonesi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Nicolò Salvarani
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- Institute of Genetics and Biomedical Research, National Research Council of Italy (Milan Unit), Rozzano (MI), Italy (N.S., P.C., C. Peano)
| | - Pierluigi Carullo
- Institute of Genetics and Biomedical Research, National Research Council of Italy (Milan Unit), Rozzano (MI), Italy (N.S., P.C., C. Peano)
| | - Federico Colombo
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Alessandra Idini
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Mauro Passaretti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Riccardo Doro
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Marcello Rubino
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Alessandro Villaschi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Giorgio Da Rin
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Clelia Peano
- Institute of Genetics and Biomedical Research, National Research Council of Italy (Milan Unit), Rozzano (MI), Italy (N.S., P.C., C. Peano)
| | - Marinos Kallikourdis
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Carolina M Greco
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Gianluigi Condorelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| |
Collapse
|
4
|
Ugarte JP, Tobón C. Fractional-order modeling of myocardium structure effects on atrial fibrillation electrograms. Math Biosci 2024; 378:109331. [PMID: 39481642 DOI: 10.1016/j.mbs.2024.109331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024]
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia with mechanisms of initiation and sustaining that are not fully understood. The clinical procedure for AF contemplates the analysis of the atrial electrograms, whose morphology has been correlated with the underlying structure of the atrial myocardium. This study employs a mathematical model incorporating fractional calculus to simulate cardiac electrical conduction, accounting for tissue structural inhomogeneities using complex-valued orders. Simulations of different wavefront propagation patterns were performed, and virtual electrograms were analyzed using an asymmetry factor. Our results evinced that the shapes of the action potential and the propagating wavefront can be modulated through the fractional order under both healthy and AF conditions. Moreover, the asymmetry factor changes with variations in the fractional order. For a given propagation pattern under AF conditions, variation intervals for the asymmetry factor can be generated by forming sets of simulations with different configurations for the fractional order, representing diverse samples of atrial tissue with varying degrees of structural heterogeneity. This approach successfully reproduces the electrogram negative deflection predominance seen in AF patients, which standard integer-order models cannot predict. Our fractional-order conduction model aligns with the effects of atrial structure on the electrical dynamics observed in clinical AF. Therefore, it offers a valuable tool for studying cardiac electrophysiology, encompassing both electrical and structural interactions of the tissue within a unified model.
Collapse
Affiliation(s)
- Juan P Ugarte
- GIMSC, Universidad de San Buenaventura, Medellin, Colombia.
| | | |
Collapse
|
5
|
Aradhyula V, Vyas R, Dube P, Haller ST, Gupta R, Maddipati KR, Kennedy DJ, Khouri SJ. Novel insights into the pathobiology of pulmonary hypertension in heart failure with preserved ejection fraction. Am J Physiol Heart Circ Physiol 2024; 326:H1498-H1514. [PMID: 38639739 PMCID: PMC11380948 DOI: 10.1152/ajpheart.00068.2024] [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: 02/02/2024] [Revised: 04/01/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is the most common cause of pulmonary hypertension (PH) worldwide and is strongly associated with adverse clinical outcomes. The American Heart Association recently highlighted a call to action regarding the distinct lack of evidence-based treatments for PH due to poorly understood pathophysiology of PH attributable to HFpEF (PH-HFpEF). Prior studies have described cardiophysiological mechanisms to explain the development of isolated postcapillary PH (ipc-PH); however, the consequent increase in pulmonary vascular (PV) resistance (PVR) may lead to the less understood and more fatal combined pre- and postcapillary PH (cpc-PH). Metabolic disease and inflammatory dysregulation have been suggested to predispose PH, yet the molecular mechanisms are unknown. Although PH-HFpEF has been studied to partly share vasoactive neurohormonal mediators with primary pulmonary arterial hypertension (PAH), clinical trials that have targeted these pathways have been unsuccessful. The increased mortality of patients with PH-HFpEF necessitates further study into viable mechanistic targets involved in disease progression. We aim to summarize the current pathophysiological and clinical understanding of PH-HFpEF, highlight the role of known molecular mechanisms in the progression of PV disease, and introduce a novel concept that lipid metabolism may be attenuating and propagating PH-HFpEF.NEW & NOTEWORTHY Our review addresses pulmonary hypertension (PH) attributable to heart failure (HF) with preserved ejection fraction (HFpEF; PH-HFpEF). Current knowledge gaps in PH-HFpEF pathophysiology have led to a lack of therapeutic targets. Thus, we address identified knowledge gaps in a comprehensive review, focusing on current clinical epidemiology, known pathophysiology, and previously studied molecular mechanisms. We also introduce a comprehensive review of polyunsaturated fatty acid (PUFA) lipid inflammatory mediators in PH-HFpEF.
Collapse
Affiliation(s)
- Vaishnavi Aradhyula
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, United States
| | - Rohit Vyas
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, United States
| | - Prabhatchandra Dube
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, United States
| | - Steven T Haller
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, United States
| | - Rajesh Gupta
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, United States
| | - Krishna Rao Maddipati
- Department of Pathology, Lipidomics Core Facility, Wayne State University, Detroit, Michigan, United States
| | - David J Kennedy
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, United States
| | - Samer J Khouri
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, United States
| |
Collapse
|
6
|
Yao J, Chen Y, Huang Y, Sun X, Shi X. The role of cardiac microenvironment in cardiovascular diseases: implications for therapy. Hum Cell 2024; 37:607-624. [PMID: 38498133 DOI: 10.1007/s13577-024-01052-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/25/2024] [Indexed: 03/20/2024]
Abstract
Due to aging populations and changes in lifestyle, cardiovascular diseases including cardiomyopathy, hypertension, and atherosclerosis, are the leading causes of death worldwide. The heart is a complicated organ composed of multicellular types, including cardiomyocytes, fibroblasts, endothelial cells, vascular smooth muscle cells, and immune cells. Cellular specialization and complex interplay between different cell types are crucial for the cardiac tissue homeostasis and coordinated function of the heart. Mounting studies have demonstrated that dysfunctional cells and disordered cardiac microenvironment are closely associated with the pathogenesis of various cardiovascular diseases. In this paper, we discuss the composition and the homeostasis of cardiac tissues, and focus on the role of cardiac environment and underlying molecular mechanisms in various cardiovascular diseases. Besides, we elucidate the novel treatment for cardiovascular diseases, including stem cell therapy and targeted therapy. Clarification of these issues may provide novel insights into the prevention and potential targets for cardiovascular diseases.
Collapse
Affiliation(s)
- Jiayu Yao
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yuejun Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yuqing Huang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| | - Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.
| |
Collapse
|
7
|
Lv L, Chen Q, Lu J, Zhao Q, Wang H, Li J, Yuan K, Dong Z. Potential regulatory role of epigenetic modifications in aging-related heart failure. Int J Cardiol 2024; 401:131858. [PMID: 38360101 DOI: 10.1016/j.ijcard.2024.131858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/27/2023] [Accepted: 02/10/2024] [Indexed: 02/17/2024]
Abstract
Heart failure (HF) is a serious clinical syndrome and a serious development or advanced stage of various heart diseases. Aging is an independent factor that causes pathological damage in cardiomyopathy and participates in the occurrence of HF at the molecular level by affecting mechanisms such as telomere shortening and mitochondrial dysfunction. Epigenetic changes have a significant impact on the aging process, and there is increasing evidence that genetic and epigenetic changes are key features of aging and aging-related diseases. Epigenetic modifications can affect genetic information by changing the chromatin state without changing the DNA sequence. Most of the genetic loci that are highly associated with cardiovascular diseases (CVD) are located in non-coding regions of the genome; therefore, the epigenetic mechanism of CVD has attracted much attention. In this review, we focus on the molecular mechanisms of HF during aging and epigenetic modifications mediating aging-related HF, emphasizing that epigenetic mechanisms play an important role in the pathogenesis of aging-related CVD and can be used as potential diagnostic and prognostic biomarkers, as well as therapeutic targets.
Collapse
Affiliation(s)
- Lin Lv
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - QiuYu Chen
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jing Lu
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qi Zhao
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - HongYan Wang
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - JiaHao Li
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - KeYing Yuan
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - ZengXiang Dong
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, First Affiliated Hospital of Harbin Medical University, Harbin, China; NHC Key Laboratory of Cell Transplantation, First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
8
|
Vijayakumar A, Wang M, Kailasam S. The Senescent Heart-"Age Doth Wither Its Infinite Variety". Int J Mol Sci 2024; 25:3581. [PMID: 38612393 PMCID: PMC11011282 DOI: 10.3390/ijms25073581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/10/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
Cardiovascular diseases are a leading cause of morbidity and mortality world-wide. While many factors like smoking, hypertension, diabetes, dyslipidaemia, a sedentary lifestyle, and genetic factors can predispose to cardiovascular diseases, the natural process of aging is by itself a major determinant of the risk. Cardiac aging is marked by a conglomerate of cellular and molecular changes, exacerbated by age-driven decline in cardiac regeneration capacity. Although the phenotypes of cardiac aging are well characterised, the underlying molecular mechanisms are far less explored. Recent advances unequivocally link cardiovascular aging to the dysregulation of critical signalling pathways in cardiac fibroblasts, which compromises the critical role of these cells in maintaining the structural and functional integrity of the myocardium. Clearly, the identification of cardiac fibroblast-specific factors and mechanisms that regulate cardiac fibroblast function in the senescent myocardium is of immense importance. In this regard, recent studies show that Discoidin domain receptor 2 (DDR2), a collagen-activated receptor tyrosine kinase predominantly located in cardiac fibroblasts, has an obligate role in cardiac fibroblast function and cardiovascular fibrosis. Incisive studies on the molecular basis of cardiovascular aging and dysregulated fibroblast function in the senescent heart would pave the way for effective strategies to mitigate cardiovascular diseases in a rapidly growing elderly population.
Collapse
Affiliation(s)
- Anupama Vijayakumar
- Cardiovascular Genetics Laboratory, Department of Biotechnology, Bhupat and Jyothi Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India;
| | - Mingyi Wang
- Laboratory of Cardiovascular Science, National Institute on Aging/National Institutes of Health, Baltimore, MD 21224, USA;
| | - Shivakumar Kailasam
- Department of Biotechnology, University of Kerala, Kariavattom, Trivandrum 695581, India
| |
Collapse
|
9
|
Song B, Zhang H, Zhou B. RETRACTED ARTICLE: Effects of Dapagliflozin on myocardial remodeling, inflammatory factors, and cardiac events in heart failure with preserved ejection fraction. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:629. [PMID: 37368031 DOI: 10.1007/s00210-023-02590-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 06/17/2023] [Indexed: 06/28/2023]
Affiliation(s)
- BoFan Song
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Shushan District, Anhui, 230022, Hefei, People's Republic of China
| | - Hao Zhang
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Shushan District, Anhui, 230022, Hefei, People's Republic of China
| | - Birong Zhou
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Shushan District, Anhui, 230022, Hefei, People's Republic of China.
| |
Collapse
|
10
|
Ahn S, Yoon JY, Kim P. Intravital imaging of cardiac tissue utilizing tissue-stabilized heart window chamber in live animal model. EUROPEAN HEART JOURNAL. IMAGING METHODS AND PRACTICE 2024; 2:qyae062. [PMID: 39224098 PMCID: PMC11367956 DOI: 10.1093/ehjimp/qyae062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/07/2024] [Indexed: 09/04/2024]
Abstract
Aims To develop and validate an optimized intravital heart microimaging protocol using a suction-based tissue motion-stabilizing cardiac imaging window to facilitate real-time observation of dynamic cellular behaviours within cardiac tissue in live mouse models. Methods and results Intravital heart imaging was conducted using dual-mode confocal and two-photon microscopy. Mice were anesthetized, intubated, and maintained at a stable body temperature during the procedure. LysM-eGFP transgenic mice were utilized to visualize immune cell dynamics with vascular labelling by intravenous injection of anti-CD31 antibody and DiD-labelled red blood cells (RBCs). A heart imaging window chamber with a vacuum-based tissue motion stabilizer with 890-920 mbar was applied following a chest incision to expose the cardiac tissue. The suction-based heart imaging window chamber system and artificial intelligence-based motion compensation function significantly reduced motion artefacts and facilitated real-time in vivo cell analysis of immune cell and RBC trafficking, revealing a mean neutrophil movement velocity of 1.66 mm/s, which was slower compared to the RBC flow velocity of 9.22 mm/s. Intravital two-photon microscopic heart imaging enabled label-free second harmonic generation imaging of cardiac muscle structures with 820-840 nm excitation wavelength, revealing detailed biodistributions and structural variations in sarcomeres and fibrillar organization in the heart. Conclusion The optimized intravital heart imaging protocol successfully demonstrates its capability to provide high-resolution, real-time visualization of dynamic cellular activities within live cardiac tissue.
Collapse
Affiliation(s)
- Soyeon Ahn
- R&D Center, IVIM Technology, 17 Techno 4-ro, Yuseong-gu, Daejeon, 34013, Republic of Korea
| | - Jung-yeon Yoon
- R&D Center, IVIM Technology, 17 Techno 4-ro, Yuseong-gu, Daejeon, 34013, Republic of Korea
| | - Pilhan Kim
- R&D Center, IVIM Technology, 17 Techno 4-ro, Yuseong-gu, Daejeon, 34013, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| |
Collapse
|
11
|
Shafi O, Siddiqui G, Jaffry HA. The benign nature and rare occurrence of cardiac myxoma as a possible consequence of the limited cardiac proliferative/ regenerative potential: a systematic review. BMC Cancer 2023; 23:1245. [PMID: 38110859 PMCID: PMC10726542 DOI: 10.1186/s12885-023-11723-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/05/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Cardiac Myxoma is a primary tumor of heart. Its origins, rarity of the occurrence of primary cardiac tumors and how it may be related to limited cardiac regenerative potential, are not yet entirely known. This study investigates the key cardiac genes/ transcription factors (TFs) and signaling pathways to understand these important questions. METHODS Databases including PubMed, MEDLINE, and Google Scholar were searched for published articles without any date restrictions, involving cardiac myxoma, cardiac genes/TFs/signaling pathways and their roles in cardiogenesis, proliferation, differentiation, key interactions and tumorigenesis, with focus on cardiomyocytes. RESULTS The cardiac genetic landscape is governed by a very tight control between proliferation and differentiation-related genes/TFs/pathways. Cardiac myxoma originates possibly as a consequence of dysregulations in the gene expression of differentiation regulators including Tbx5, GATA4, HAND1/2, MYOCD, HOPX, BMPs. Such dysregulations switch the expression of cardiomyocytes into progenitor-like state in cardiac myxoma development by dysregulating Isl1, Baf60 complex, Wnt, FGF, Notch, Mef2c and others. The Nkx2-5 and MSX2 contribute predominantly to both proliferation and differentiation of Cardiac Progenitor Cells (CPCs), may possibly serve roles based on the microenvironment and the direction of cell circuitry in cardiac tumorigenesis. The Nkx2-5 in cardiac myxoma may serve to limit progression of tumorigenesis as it has massive control over the proliferation of CPCs. The cardiac cell type-specific genetic programming plays governing role in controlling the tumorigenesis and regenerative potential. CONCLUSION The cardiomyocytes have very limited proliferative and regenerative potential. They survive for long periods of time and tightly maintain the gene expression of differentiation genes such as Tbx5, GATA4 that interact with tumor suppressors (TS) and exert TS like effect. The total effect such gene expression exerts is responsible for the rare occurrence and benign nature of primary cardiac tumors. This prevents the progression of tumorigenesis. But this also limits the regenerative and proliferative potential of cardiomyocytes. Cardiac Myxoma develops as a consequence of dysregulations in these key genes which revert the cells towards progenitor-like state, hallmark of CM. The CM development in carney complex also signifies the role of TS in cardiac cells.
Collapse
Affiliation(s)
- Ovais Shafi
- Sindh Medical College - Jinnah Sindh Medical University / Dow University of Health Sciences, Karachi, Pakistan.
| | - Ghazia Siddiqui
- Sindh Medical College - Jinnah Sindh Medical University / Dow University of Health Sciences, Karachi, Pakistan
| | - Hassam A Jaffry
- Sindh Medical College - Jinnah Sindh Medical University / Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
12
|
Li J, Xin Y, Wang Z, Li J, Li W, Li H. The role of cardiac resident macrophage in cardiac aging. Aging Cell 2023; 22:e14008. [PMID: 37817547 PMCID: PMC10726886 DOI: 10.1111/acel.14008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/12/2023] Open
Abstract
Advancements in longevity research have provided insights into the impact of cardiac aging on the structural and functional aspects of the heart. Notable changes include the gradual remodeling of the myocardium, the occurrence of left ventricular hypertrophy, and the decline in both systolic and diastolic functions. Macrophages, a type of immune cell, play a pivotal role in innate immunity by serving as vigilant agents against pathogens, facilitating wound healing, and orchestrating the development of targeted acquired immune responses. Distinct subsets of macrophages are present within the cardiac tissue and demonstrate varied functions in response to myocardial injury. The differentiation of cardiac macrophages according to their developmental origin has proven to be a valuable strategy in identifying reparative macrophage populations, which originate from embryonic cells and reside within the tissue, as well as inflammatory macrophages, which are derived from monocytes and recruited to the heart. These subsets of macrophages possess unique characteristics and perform distinct functions. This review aims to summarize the current understanding of the roles and phenotypes of cardiac macrophages in various conditions, including the steady state, aging, and other pathological conditions. Additionally, it will highlight areas that require further investigation to expand our knowledge in this field.
Collapse
Affiliation(s)
- Jiayu Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
- Laboratory for Clinical MedicineBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Yanguo Xin
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
- Laboratory for Clinical MedicineBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Zhaojia Wang
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
- Laboratory for Clinical MedicineBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Jingye Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Weiping Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
- Laboratory for Clinical MedicineBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Hongwei Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
- Laboratory for Clinical MedicineBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
- Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular DiseaseBeijingChina
| |
Collapse
|
13
|
Xu ST, Zhang YX, Liu SL, Liu F, Ye JT. Exosomes derived from cardiac fibroblasts with angiotensin II stimulation provoke hypertrophy and autophagy inhibition in cardiomyocytes. Biochem Biophys Res Commun 2023; 682:199-206. [PMID: 37826943 DOI: 10.1016/j.bbrc.2023.10.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 10/07/2023] [Indexed: 10/14/2023]
Abstract
Although accumulating evidence has revealed that autophagy inhibition contributes to the development of pathological cardiac hypertrophy, the mechanisms leading to declined autophagy activity in the hypertrophic heart remain to be elucidated. Exosomes are known to be important mediators of intercellular communication, and the involvement of exosomes in cardiovascular abnormities has attracted increasing attentions. Cardiac fibroblasts (CFs) are the most abundant cell type in the heart. Here, we investigated the potential role of CFs-derived exosomes in regulating cardiomyocyte hypertrophy and autophagy. Exosomes from rat CFs treated with angiotensin II (Ang II-CFs-exosomes) were collected and characterized. Our experiments showed that these exosomes could induce hypertrophic responses and impair autophagy activity in primary neonatal rat cardiomyocytes (NRCMs). Ang II-CFs-exosomes blocked the autophagic flux of NRCMs via inhibiting the formation of autolysosomes. Moreover, the pro-hypertrophic effects and autophagy inhibition induced by Ang II-CFs-exosomes was validated in mice receiving injection of the exosomes. These findings highlight a novel role of Ang II-CFs-exosomes in suppressing cardiomyocyte autophagy, which may help to better understand the pathogenesis of cardiac hypertrophy.
Collapse
Affiliation(s)
- Si-Ting Xu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangzhou, 510006, China
| | - Yue-Xin Zhang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangzhou, 510006, China
| | - Si-Ling Liu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangzhou, 510006, China
| | - Fang Liu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangzhou, 510006, China
| | - Jian-Tao Ye
- School of Pharmaceutical Sciences, Sun Yat-Sen University, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangzhou, 510006, China.
| |
Collapse
|
14
|
Windt LM, Wiendels M, Dostanić M, Bellin M, Sarro PM, Mastrangeli M, Mummery CL, van Meer BJ. Miniaturized engineered heart tissues from hiPSC-derived triple cell type co-cultures to study human cardiac function. Biochem Biophys Res Commun 2023; 681:200-211. [PMID: 37783118 DOI: 10.1016/j.bbrc.2023.09.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/23/2023] [Accepted: 09/14/2023] [Indexed: 10/04/2023]
Abstract
Human heart tissues grown as three-dimensional spheroids and consisting of different cardiac cell types derived from pluripotent stem cells (hiPSCs) recapitulate aspects of human physiology better than standard two-dimensional models in vitro. They typically consist of less than 5000 cells and are used to measure contraction kinetics although not contraction force. By contrast, engineered heart tissues (EHTs) formed around two flexible pillars, can measure contraction force but conventional EHTs often require between 0.5 and 2 million cells. This makes large-scale screening of many EHTs costly. Our goals here were (i) to create a physiologically relevant model that required fewer cells than standard EHTs making them less expensive, and (ii) to ensure that this miniaturized model retained correct functionality. We demonstrated that fully functional EHTs could be generated from physiologically relevant combinations of hiPSC-derived cardiomyocytes (70%), cardiac fibroblasts (15%) and cardiac endothelial cells (15%), using as few as 1.6 × 104 cells. Our results showed that these EHTs were viable and functional up to 14 days after formation. The EHTs could be electrically paced in the frequency range between 0.6 and 3 Hz, with the optimum between 0.6 and 2 Hz. This was consistent across three downscaled EHT sizes tested. These findings suggest that miniaturized EHTs could represent a cost-effective microphysiological system for disease modelling and examining drug responses particularly in secondary screens for drug discovery.
Collapse
Affiliation(s)
- L M Windt
- Department of Anatomy and Embryology, LUMC, Leiden, the Netherlands
| | - M Wiendels
- Department of Anatomy and Embryology, LUMC, Leiden, the Netherlands
| | - M Dostanić
- Department of Anatomy and Embryology, LUMC, Leiden, the Netherlands; Microelectronics, TU Delft, Delft, the Netherlands
| | - M Bellin
- Department of Anatomy and Embryology, LUMC, Leiden, the Netherlands; Department of Biology, University of Padua, Padua, Italy; Veneto Institute of Molecular Medicine, Padua, Italy
| | - P M Sarro
- Microelectronics, TU Delft, Delft, the Netherlands
| | | | - C L Mummery
- Department of Anatomy and Embryology, LUMC, Leiden, the Netherlands
| | - B J van Meer
- Department of Anatomy and Embryology, LUMC, Leiden, the Netherlands; Sync Biosystems, Leiden, the Netherlands.
| |
Collapse
|
15
|
Meng XM, Liu SB, Deng T, Li DY, You L, Hong H, Feng QP, Zhu BM. Loss of Histone Methyltransferase KMT2D Attenuates Angiogenesis in the Ischemic Heart by Inhibiting the Transcriptional Activation of VEGF-A. J Cardiovasc Transl Res 2023; 16:1032-1049. [PMID: 36947365 PMCID: PMC10616223 DOI: 10.1007/s12265-023-10373-x] [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: 02/02/2023] [Accepted: 03/08/2023] [Indexed: 03/23/2023]
Abstract
Angiogenesis occurred after myocardial infarction (MI) protects heart failure (HF). The aim of our study was to explore function of histone methyltransferase KMT2D (MLL4, mixed-lineage leukemia 4) in angiogenesis post-MI. Western blotting showed that KMT2D protein expression was elevated in MI mouse myocardial. Cardiomyocyte-specific Kmt2d-knockout (Kmt2d-cKO) mice were generated, and echocardiography and immunofluorescence staining detected significantly attenuated cardiac function and insufficient angiogenesis following MI in Kmt2d-cKO mice. Cross-talk assay suggested that Kmt2d-KO H9c2-derived conditioned medium attenuates EA.hy926 EC function. ELISA further identified that VEGF-A released from Kmt2d-KO H9c2 was significantly reduced. CUT&Tag and RT-qPCR revealed that KMT2D deficiency reduced Vegf-a mRNA expression and enrichment of H3K4me1 on the Vegf-a promoter. Moreover, KMT2D silencing in ECs also suppressed endothelial function. Our study indicates that KMT2D depletion in both cardiomyocytes and ECs attenuates angiogenesis and that loss of KMT2D exacerbates heart failure after MI in mice.
Collapse
Affiliation(s)
- Xiang-Min Meng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shu-Bao Liu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tian Deng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - De-Yong Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lu You
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hao Hong
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qi-Pu Feng
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bing-Mei Zhu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
16
|
Lother A, Kohl P. The heterocellular heart: identities, interactions, and implications for cardiology. Basic Res Cardiol 2023; 118:30. [PMID: 37495826 PMCID: PMC10371928 DOI: 10.1007/s00395-023-01000-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/17/2023] [Accepted: 07/17/2023] [Indexed: 07/28/2023]
Abstract
The heterocellular nature of the heart has been receiving increasing attention in recent years. In addition to cardiomyocytes as the prototypical cell type of the heart, non-myocytes such as endothelial cells, fibroblasts, or immune cells are coming more into focus. The rise of single-cell sequencing technologies enables identification of ever more subtle differences and has reignited the question of what defines a cell's identity. Here we provide an overview of the major cardiac cell types, describe their roles in homeostasis, and outline recent findings on non-canonical functions that may be of relevance for cardiology. We highlight modes of biochemical and biophysical interactions between different cardiac cell types and discuss the potential implications of the heterocellular nature of the heart for basic research and therapeutic interventions.
Collapse
Affiliation(s)
- Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstr. 25, 79104, Freiburg, Germany.
- Interdisciplinary Medical Intensive Care, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany.
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, Faculty of Medicine, University Heart Center, University of Freiburg, Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW The intricate interplay between inflammatory and reparative responses in the context of heart injury is central to the pathogenesis of heart failure. Recent clinical studies have shown the therapeutic benefits of anti-inflammatory strategies in the treatment of cardiovascular diseases. This review provides a comprehensive overview of the cross-talk between immune cells and fibroblasts in the diseased heart. RECENT FINDINGS The role of inflammatory cells in fibroblast activation after cardiac injury is well-documented, but recent single-cell transcriptomics studies have identified putative pro-inflammatory fibroblasts in the infarcted heart, suggesting that fibroblasts, in turn, can modify inflammatory cell behavior. Furthermore, anti-inflammatory immune cells and fibroblasts have been described. The use of spatial and temporal-omics analyses may provide additional insights toward a better understanding of disease-specific microenvironments, where activated fibroblasts and inflammatory cells are in proximity. Recent studies focused on the interplay between fibroblasts and immune cells have brought us closer to the identification of cell type-specific targets for intervention. Further exploration of these intercellular communications will provide deeper insights toward the development of novel therapeutics.
Collapse
Affiliation(s)
- Akitoshi Hara
- Center for Cardiovascular Research, University of Hawaii at Manoa, Honolulu, HI, 96825, USA.
| | - Michelle D Tallquist
- Center for Cardiovascular Research, University of Hawaii at Manoa, Honolulu, HI, 96825, USA
| |
Collapse
|
18
|
Cuenca-Bermejo L, Fernández-Del Palacio MJ, de Cassia Gonçalves V, Bautista-Hernández V, Sánchez-Rodrigo C, Fernández-Villalba E, Kublickiene K, Raparelli V, Kautzky-Willer A, Norris CM, Pilote L, Herrero MT. Age and Sex Determine Electrocardiogram Parameters in the Octodon degus. BIOLOGY 2023; 12:747. [PMID: 37237559 PMCID: PMC10215068 DOI: 10.3390/biology12050747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/02/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023]
Abstract
Cardiovascular diseases represent the leading cause of mortality and morbidity worldwide, and age is an important risk factor. Preclinical models provide supportive evidence toward age-related cardiac changes, as well as allow for the study of pathological aspects of the disease. In the present work, we evaluated the electrocardiogram (ECG) recording in the O. degus during the aging process in both females and males. Taking into account the age and sex, our study provides the normal ranges for the heart rate, duration and voltage of the ECG waves and intervals, as well as electrical axis deviation. We found that the QRS complex duration and QTc significantly increased with age, whereas the heart rate significantly decreased. On the other hand, the P wave, PR and QTc segments durations, S wave voltage and electrical axis were found to be significantly different between males and females. The heart rhythm was also altered in aged animals, resulting in an increased incidence of arrhythmias, especially in males. Based on these results, we suggest that this rodent model could be useful for cardiovascular research, including impacts of aging and biological sex.
Collapse
Affiliation(s)
- Lorena Cuenca-Bermejo
- Clinical & Experimental Neuroscience (NiCE), Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), University of Murcia, 30120 Murcia, Spain; (L.C.-B.); (V.d.C.G.); (C.S.-R.); (E.F.-V.)
- Institute for Aging Research (IUIE), Campus Mare Nostrum, European University for Wellbeing (EUniWell), 30100 Murcia, Spain
| | | | - Valeria de Cassia Gonçalves
- Clinical & Experimental Neuroscience (NiCE), Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), University of Murcia, 30120 Murcia, Spain; (L.C.-B.); (V.d.C.G.); (C.S.-R.); (E.F.-V.)
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Universidade Federal de São Paulo (UNIFESP), São Paulo 04039-032, Brazil
| | | | - Consuelo Sánchez-Rodrigo
- Clinical & Experimental Neuroscience (NiCE), Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), University of Murcia, 30120 Murcia, Spain; (L.C.-B.); (V.d.C.G.); (C.S.-R.); (E.F.-V.)
- Institute for Aging Research (IUIE), Campus Mare Nostrum, European University for Wellbeing (EUniWell), 30100 Murcia, Spain
| | - Emiliano Fernández-Villalba
- Clinical & Experimental Neuroscience (NiCE), Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), University of Murcia, 30120 Murcia, Spain; (L.C.-B.); (V.d.C.G.); (C.S.-R.); (E.F.-V.)
- Institute for Aging Research (IUIE), Campus Mare Nostrum, European University for Wellbeing (EUniWell), 30100 Murcia, Spain
| | - Karolina Kublickiene
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17177 Stockholm, Sweden;
| | - Valeria Raparelli
- Department of Translational Medicine, University of Ferrara, 44124 Ferrara, Italy;
- University Center for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Alexandra Kautzky-Willer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria;
| | - Colleen M. Norris
- Faculty of Nursing, University of Alberta, Edmonton, AB T6G 2R3, Canada;
- Cardiovascular and Stroke Strategic Clinical Network, Alberta Health Services, Edmonton, AB T5J 3E4, Canada
| | - Louise Pilote
- Division of Clinical Epidemiology, Research Institute of McGill University Health Centre, McGill University, Montreal, QC H4A 3J1, Canada;
| | - María Trinidad Herrero
- Clinical & Experimental Neuroscience (NiCE), Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), University of Murcia, 30120 Murcia, Spain; (L.C.-B.); (V.d.C.G.); (C.S.-R.); (E.F.-V.)
- Institute for Aging Research (IUIE), Campus Mare Nostrum, European University for Wellbeing (EUniWell), 30100 Murcia, Spain
| |
Collapse
|
19
|
Varghese LN, Schwenke DO, Katare R. Role of noncoding RNAs in cardiac ageing. Front Cardiovasc Med 2023; 10:1142575. [PMID: 37034355 PMCID: PMC10073704 DOI: 10.3389/fcvm.2023.1142575] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
The global population is estimated to reach 9.8 billion by 2050, of which 2.1 billion will comprise individuals above 60 years of age. As the number of elderly is estimated to double from 2017, it is a victory of the modern healthcare system but also worrisome as ageing, and the onset of chronic disease are correlated. Among other chronic conditions, cardiovascular diseases (CVDs) are the leading cause of death in the aged population. While the underlying cause of the age-associated development of CVDs is not fully understood, studies indicate the role of non-coding RNAs such as microRNAs (miRNAs) and long noncoding RNAs (lnc-RNAs) in the development of age-associated CVDs. miRNAs and lnc-RNAs are non-coding RNAs which control gene expression at the post-transcriptional level. The expression of specific miRNAs and lnc-RNAs are reportedly dysregulated with age, leading to cardiovascular system changes and ultimately causing CVDs. Since miRNAs and lnc-RNAs play several vital roles in maintaining the normal functioning of the cardiovascular system, they are also being explored for their therapeutic potential as a treatment for CVDs. This review will first explore the pathophysiological changes associated with ageing. Next, we will review the known mechanisms underlying the development of CVD in ageing with a specific focus on miRNA and lnc-RNAs. Finally, we will discuss the therapeutic options and future challenges towards healthy cardiac ageing. With the global ageing population on the rise, this review will provide a fundamental understanding of some of the underlying molecular mechanisms of cardiac ageing.
Collapse
Affiliation(s)
| | | | - Rajesh Katare
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
20
|
Li Q, Xiao M, Li N, Cai W, Zhao C, Liu B, Zeng F. Application of
Caenorhabditis elegans
in the evaluation of food nutrition: A review. EFOOD 2023. [DOI: 10.1002/efd2.68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Affiliation(s)
- Quancen Li
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Meifang Xiao
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Na Li
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Wenwen Cai
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Chao Zhao
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- Engineering Research Center of Fujian Subtropical Fruit and Vegetable Processing Fujian Agriculture and Forestry University Fuzhou China
| | - Bin Liu
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
- Engineering Research Center of Fujian Subtropical Fruit and Vegetable Processing Fujian Agriculture and Forestry University Fuzhou China
- National Engineering Research Center of JUNCAO Technology Fujian Agriculture and Forestry University Fuzhou China
| | - Feng Zeng
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
- Engineering Research Center of Fujian Subtropical Fruit and Vegetable Processing Fujian Agriculture and Forestry University Fuzhou China
| |
Collapse
|
21
|
Exosome Release by Glucose Deprivation Is Important for the Viability of TSC-Null Cells. Cells 2022; 11:cells11182862. [PMID: 36139445 PMCID: PMC9497210 DOI: 10.3390/cells11182862] [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: 07/22/2022] [Revised: 09/05/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
The control of exosome release is associated with numerous physiological and pathological activities, and that release is often indicative of health, disease, and environmental nutrient stress. Tuberous sclerosis complex (TSC) regulates the cell viability via the negative regulation of the mammalian target of rapamycin complex (mTORC1) during glucose deprivation. However, the mechanism by which viability of TSC-null cells is regulated by mTORC1 inhibition under glucose deprivation remains unclear. Here, we demonstrated that exosome release regulates cell death induced by glucose deprivation in TSC-null cells. The mTORC1 inhibition by rapamycin significantly increased the exosome biogenesis, exosome secretion, and cell viability in TSC-null cells. In addition, the increase in cell viability by mTORC1 inhibition was attenuated by two different types of inhibitors of exosome release under glucose deprivation. Taken together, we suggest that exosome release inhibition might be a novel way for regression of cell growth in TSC-null cells showing lack of cell death by mTORC1 inhibition.
Collapse
|
22
|
Wang Y, Wei J, Zhang P, Zhang X, Wang Y, Chen W, Zhao Y, Cui X. Neuregulin-1, a potential therapeutic target for cardiac repair. Front Pharmacol 2022; 13:945206. [PMID: 36120374 PMCID: PMC9471952 DOI: 10.3389/fphar.2022.945206] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
NRG1 (Neuregulin-1) is an effective cardiomyocyte proliferator, secreted and released by endothelial vascular cells, and affects the cardiovascular system. It plays a major role in heart growth, proliferation, differentiation, apoptosis, and other cardiovascular processes. Numerous experiments have shown that NRG1 can repair the heart in the pathophysiology of atherosclerosis, myocardial infarction, ischemia reperfusion, heart failure, cardiomyopathy and other cardiovascular diseases. NRG1 can connect related signaling pathways through the NRG1/ErbB pathway, which form signal cascades to improve the myocardial microenvironment, such as regulating cardiac inflammation, oxidative stress, necrotic apoptosis. Here, we summarize recent research advances on the molecular mechanisms of NRG1, elucidate the contribution of NRG1 to cardiovascular disease, discuss therapeutic approaches targeting NRG1 associated with cardiovascular disease, and highlight areas for future research.
Collapse
Affiliation(s)
- Yan Wang
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jianliang Wei
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Peng Zhang
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xin Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yifei Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Wenjing Chen
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yanan Zhao
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- *Correspondence: Yanan Zhao, ; Xiangning Cui,
| | - Xiangning Cui
- Department of Cardiovascular, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yanan Zhao, ; Xiangning Cui,
| |
Collapse
|
23
|
Liang Z, Miao Y, Teng X, Xiao L, Guo Q, Xue H, Tian D, Jin S, Wu Y. Hydrogen Sulfide Inhibits Ferroptosis in Cardiomyocytes to Protect Cardiac Function in Aging Rats. Front Mol Biosci 2022; 9:947778. [PMID: 35936785 PMCID: PMC9355033 DOI: 10.3389/fmolb.2022.947778] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/22/2022] [Indexed: 12/11/2022] Open
Abstract
Aging contributes significantly to cardiovascular diseases and cardiac dysfunction. To explore the reasons for the decline in cardiac function in the elderly, we collected clinical data and blood samples from 231 individuals. Our results indicated that aging was accompanied by a decline in cardiac function and remodeling of the left ventricle, and cardiac function was negatively correlated with age. Serum hydrogen sulfide (H2S) decreased, while serum malondialdehyde (MDA) and iron increased with aging in healthy individuals. A rat model of aging and iron overload was constructed for in vivo research. In the animal model, we found that the expression of endogenous H2S-producing enzymes decreased, and endogenous H2S levels decreased, while oxidative stress levels rose. The regulation of iron metabolism and the maintenance of iron homeostasis declined. The accumulation of MDA and iron led to ferroptotic cell death and subsequent myocardial injury and deterioration. A high-iron diet accelerated the aging process and death in rats. The decline of cardiac function in aging rats and iron-overload rats may be caused by cardiomyocyte ferroptosis. Exogenous H2S enhanced the expression of endogenous H2S synthase, promoted endogenous H2S production, regulated iron metabolism, and reduced oxidative stress levels. The protective effects of H2S on cardiac function in aging rats and iron-overload rats may be partly due to the inhibition of cardiomyocyte ferroptosis. We demonstrated that cardiac dysfunction associated with aging was closely related to decreased endogenous H2S levels and cardiomyocyte ferroptosis. H2S-regulated iron metabolism reduced oxidative stress levels in cardiomyocytes, inhibited cardiomyocyte ferroptosis, and protected cardiac function in aging rats.
Collapse
Affiliation(s)
- Zihui Liang
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Yuxin Miao
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Xu Teng
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Lin Xiao
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Qi Guo
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Hongmei Xue
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Danyang Tian
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Sheng Jin
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
- *Correspondence: Sheng Jin, ; Yuming Wu,
| | - Yuming Wu
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, China
- *Correspondence: Sheng Jin, ; Yuming Wu,
| |
Collapse
|
24
|
Pang L, Jiang X, Lian X, Chen J, Song EF, Jin LG, Xia ZY, Ma HC, Cai Y. Caloric restriction-mimetics for the reduction of heart failure risk in aging heart: with consideration of gender-related differences. Mil Med Res 2022; 9:33. [PMID: 35786219 PMCID: PMC9252041 DOI: 10.1186/s40779-022-00389-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 05/30/2022] [Indexed: 11/10/2022] Open
Abstract
The literature is full of claims regarding the consumption of polyphenol or polyamine-rich foods that offer some protection from developing cardiovascular disease (CVD). This is achieved by preventing cardiac hypertrophy and protecting blood vessels through improving the function of endothelium. However, do these interventions work in the aged human hearts? Cardiac aging is accompanied by an increase in left ventricular hypertrophy, along with diastolic and systolic dysfunction. It also confers significant cardiovascular risks for both sexes. The incidence and prevalence of CVD increase sharply at an earlier age in men than women. Furthermore, the patterns of heart failure differ between sexes, as do the lifetime risk factors. Do caloric restriction (CR)-mimetics, rich in polyphenol or polyamine, delay or reverse cardiac aging equally in both men and women? This review will discuss three areas: (1) mechanisms underlying age-related cardiac remodeling; (2) gender-related differences and potential mechanisms underlying diminished cardiac response in older men and women; (3) we select a few polyphenol or polyamine rich compounds as the CR-mimetics, such as resveratrol, quercetin, curcumin, epigallocatechin gallate and spermidine, due to their capability to extend health-span and induce autophagy. We outline their abilities and issues on retarding aging in animal hearts and preventing CVD in humans. We discuss the confounding factors that should be considered for developing therapeutic strategies against cardiac aging in humans.
Collapse
Affiliation(s)
- Lei Pang
- Department of Anesthesiology, the First Hospital of Jilin University, Changchun, 130021, China
| | - Xi Jiang
- Health Promotion Center, the First Hospital of Jilin University, Changchun, 130021, China
| | - Xin Lian
- Department of Urology, the First Hospital of Jilin University, Changchun, 130021, China
| | - Jie Chen
- Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan, 512000, Guangdong, China
| | - Er-Fei Song
- Department of Metabolic and Bariatric Surgery, Jinan University First Affiliated Hospital, Guangzhou, 510630, China.,Department of Medicine, LKS Faculty of Medicine, the University of Hong Kong, Hong Kong, China
| | - Lei-Gang Jin
- Department of Medicine, LKS Faculty of Medicine, the University of Hong Kong, Hong Kong, China.,State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, the University of Hong Kong, Hong Kong, China
| | - Zheng-Yuan Xia
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, the University of Hong Kong, Hong Kong, China.,Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Hai-Chun Ma
- Department of Anesthesiology, the First Hospital of Jilin University, Changchun, 130021, China.
| | - Yin Cai
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, China.
| |
Collapse
|
25
|
Abstract
It is well established that humans and other mammals are minimally regenerative compared with organisms such as zebrafish, salamander or amphibians. In recent years, however, the identification of regenerative potential in neonatal mouse tissues that normally heal poorly in adults has transformed our understanding of regenerative capacity in mammals. In this Review, we survey the mammalian tissues for which regenerative or improved neonatal healing has been established, including the heart, cochlear hair cells, the brain and spinal cord, and dense connective tissues. We also highlight common and/or tissue-specific mechanisms of neonatal regeneration, which involve cells, signaling pathways, extracellular matrix, immune cells and other factors. The identification of such common features across neonatal tissues may direct therapeutic strategies that will be broadly applicable to multiple adult tissues.
Collapse
Affiliation(s)
| | - Alice H. Huang
- Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
| |
Collapse
|
26
|
Cardiac fibroblasts regulate the development of heart failure via Htra3-TGF-β-IGFBP7 axis. Nat Commun 2022; 13:3275. [PMID: 35672400 PMCID: PMC9174232 DOI: 10.1038/s41467-022-30630-y] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 05/03/2022] [Indexed: 01/07/2023] Open
Abstract
Tissue fibrosis and organ dysfunction are hallmarks of age-related diseases including heart failure, but it remains elusive whether there is a common pathway to induce both events. Through single-cell RNA-seq, spatial transcriptomics, and genetic perturbation, we elucidate that high-temperature requirement A serine peptidase 3 (Htra3) is a critical regulator of cardiac fibrosis and heart failure by maintaining the identity of quiescent cardiac fibroblasts through degrading transforming growth factor-β (TGF-β). Pressure overload downregulates expression of Htra3 in cardiac fibroblasts and activated TGF-β signaling, which induces not only cardiac fibrosis but also heart failure through DNA damage accumulation and secretory phenotype induction in failing cardiomyocytes. Overexpression of Htra3 in the heart inhibits TGF-β signaling and ameliorates cardiac dysfunction after pressure overload. Htra3-regulated induction of spatio-temporal cardiac fibrosis and cardiomyocyte secretory phenotype are observed specifically in infarct regions after myocardial infarction. Integrative analyses of single-cardiomyocyte transcriptome and plasma proteome in human reveal that IGFBP7, which is a cytokine downstream of TGF-β and secreted from failing cardiomyocytes, is the most predictable marker of advanced heart failure. These findings highlight the roles of cardiac fibroblasts in regulating cardiomyocyte homeostasis and cardiac fibrosis through the Htra3-TGF-β-IGFBP7 pathway, which would be a therapeutic target for heart failure. Cardiac fibrosis is a hallmark of heart failure. Here the authors use single-cell RNA-sequencing, spatial transcriptomics, and genetic manipulations, to show that Htra3 regulates cardiac fibrosis by keeping fibroblasts quiescent and by degrading TGF-beta.
Collapse
|
27
|
González A, Richards AM, de Boer RA, Thum T, Arfsten H, Hülsmann M, Falcao-Pires I, Díez J, Foo RSY, Chan MY, Aimo A, Anene-Nzelu CG, Abdelhamid M, Adamopoulos S, Anker SD, Belenkov Y, Ben Gal T, Cohen-Solal A, Böhm M, Chioncel O, Delgado V, Emdin M, Jankowska EA, Gustafsson F, Hill L, Jaarsma T, Januzzi JL, Jhund PS, Lopatin Y, Lund LH, Metra M, Milicic D, Moura B, Mueller C, Mullens W, Núñez J, Piepoli MF, Rakisheva A, Ristić AD, Rossignol P, Savarese G, Tocchetti CG, Van Linthout S, Volterrani M, Seferovic P, Rosano G, Coats AJS, Bayés-Genís A. Cardiac remodelling - Part 1: From cells and tissues to circulating biomarkers. A review from the Study Group on Biomarkers of the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2022; 24:927-943. [PMID: 35334137 DOI: 10.1002/ejhf.2493] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/09/2022] [Accepted: 03/21/2022] [Indexed: 11/10/2022] Open
Abstract
Cardiac remodelling refers to changes in left ventricular structure and function over time, with a progressive deterioration that may lead to heart failure (HF) development (adverse remodelling) or vice versa a recovery (reverse remodelling) in response to HF treatment. Adverse remodelling predicts a worse outcome, whilst reverse remodelling predicts a better prognosis. The geometry, systolic and diastolic function and electric activity of the left ventricle are affected, as well as the left atrium and on the long term even right heart chambers. At a cellular and molecular level, remodelling involves all components of cardiac tissue: cardiomyocytes, fibroblasts, endothelial cells and leucocytes. The molecular, cellular and histological signatures of remodelling may differ according to the cause and severity of cardiac damage, and clearly to the global trend toward worsening or recovery. These processes cannot be routinely evaluated through endomyocardial biopsies, but may be reflected by circulating levels of several biomarkers. Different classes of biomarkers (e.g. proteins, non-coding RNAs, metabolites and/or epigenetic modifications) and several biomarkers of each class might inform on some aspects on HF development, progression and long-term outcomes, but most have failed to enter clinical practice. This may be due to the biological complexity of remodelling, so that no single biomarker could provide great insight on remodelling when assessed alone. Another possible reason is a still incomplete understanding of the role of biomarkers in the pathophysiology of cardiac remodelling. Such role will be investigated in the first part of this review paper on biomarkers of cardiac remodelling.
Collapse
Affiliation(s)
- Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, and IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - A Mark Richards
- Department of medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore
- Christchurch Heart Institute, University of Otago, Dunedin, New Zealand
| | - Rudolf A de Boer
- University Medical Center Groningen, University of Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS) and Rebirth Center for Translational Regenerative Therapies, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
| | - Henrike Arfsten
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Martin Hülsmann
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Inês Falcao-Pires
- Department od Surgery and Physiology, Cardiovascular Research and Development Center, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Javier Díez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, and IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
- Departments of Cardiology and Cardiac Surgery, and Nephrology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Roger S Y Foo
- Department of medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore
| | - Mark Y Chan
- Department of medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore
| | - Alberto Aimo
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Chukwuemeka G Anene-Nzelu
- Department of medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore
- Montreal Heart Institute, Montreal, Canada
| | | | - Stamatis Adamopoulos
- 2nd Department of Cardiovascular Medicine, Onassis Cardiac Surgery Center, Athens, Greece
| | - Stefan D Anker
- Department of Cardiology (CVK), and Berlin Institute of Health Center for Regenerative Therapies (BCRT), German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin, Berlin, Germany
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | | | - Tuvia Ben Gal
- Cardiology Department, Rabin Medical Center, Beilinson, Israel
| | | | - Michael Böhm
- Universitätsklinikum des Saarlandes, Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Saarland University, Homburg/Saar, Germany
| | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases 'Prof. C.C. Iliescu' Bucharest, University of Medicine Carol Davila, Bucharest, Romania
| | - Victoria Delgado
- Institut del Cor, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Michele Emdin
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Ewa A Jankowska
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Finn Gustafsson
- Rigshospitalet-Copenhagen University Hospital, Heart Centre, Department of Cardiology, Copenhagen, Denmark
| | | | | | - James L Januzzi
- Massachusetts General Hospital and Baim Institute for Clinical Research, Boston, MA, USA
| | - Pardeep S Jhund
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland
| | - Yuri Lopatin
- Volgograd State Medical University, Volgograd, Russia
| | - Lars H Lund
- Department of Medicine, Karolinska Institutet, and Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Marco Metra
- Cardiology, ASST Spedali Civili; Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Davor Milicic
- University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Brenda Moura
- Faculty of Medicine, University of Porto, Porto, Portugal
- Cardiology Department, Porto Armed Forces Hospital, Portugal
| | | | | | - Julio Núñez
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
- Hospital Clínico Universitario de Valencia, INCLIVA, Universidad de Valencia, Valencia, Spain
| | - Massimo F Piepoli
- Cardiology Division, Castelsangiovanni Hospital, Castelsangiovanni, Italy
| | - Amina Rakisheva
- Scientific Research Institute of Cardiology and Internal Medicine, Almaty, Kazakhstan
| | - Arsen D Ristić
- Department of Cardiology, University Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Patrick Rossignol
- Université de Lorraine, Centre d'Investigations Cliniques- Plurithématique 1433, and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Gianluigi Savarese
- Department of Medicine, Karolinska Institutet, and Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Carlo G Tocchetti
- Cardio-Oncology Unit, Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Sophie Van Linthout
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | | | - Petar Seferovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Giuseppe Rosano
- St. George's Hospitals, NHS Trust, University of London, London, UK
| | | | - Antoni Bayés-Genís
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
- Institut del Cor, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
28
|
Gou B, Chu X, Xiao Y, Liu P, Zhang H, Gao Z, Song M. Single-Cell Analysis Reveals Transcriptomic Reprogramming in Aging Cardiovascular Endothelial Cells. Front Cardiovasc Med 2022; 9:900978. [PMID: 35615560 PMCID: PMC9124831 DOI: 10.3389/fcvm.2022.900978] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/04/2022] [Indexed: 11/18/2022] Open
Abstract
The senescence of cardiovascular endothelial cells (ECs) is a major risk factor in the development of aging-related cardiovascular diseases. However, the molecular dynamics in cardiovascular EC aging are poorly understood. Here, we characterized the transcriptomic landscape of cardiovascular ECs during aging and observed that ribosome biogenesis, inflammation, apoptosis and angiogenesis-related genes and pathways changed with age. We also highlighted the importance of collagen genes in the crosstalk between ECs and other cell types in cardiovascular aging. Moreover, transcriptional regulatory network analysis revealed Jun as a candidate transcription factor involved in murine cardiovascular senescence and we validated the upregulation of Jun in aged cardiovascular ECs both in vitro and in vivo. Altogether, our study reveals the transcriptomic reprogramming in the aging murine cardiovascular ECs, which deepens the understanding of the molecular mechanisms of cardiovascular aging and provides new insights into potential therapeutic targets against age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Bo Gou
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaojing Chu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Yi Xiao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Pinxuan Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hao Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zeyu Gao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
29
|
Besse S, Nadaud S, Balse E, Pavoine C. Early Protective Role of Inflammation in Cardiac Remodeling and Heart Failure: Focus on TNFα and Resident Macrophages. Cells 2022; 11:1249. [PMID: 35406812 PMCID: PMC8998130 DOI: 10.3390/cells11071249] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 02/24/2022] [Accepted: 04/01/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiac hypertrophy, initiated by a variety of physiological or pathological stimuli (hemodynamic or hormonal stimulation or infarction), is a critical early adaptive compensatory response of the heart. The structural basis of the progression from compensated hypertrophy to pathological hypertrophy and heart failure is still largely unknown. In most cases, early activation of an inflammatory program reflects a reparative or protective response to other primary injurious processes. Later on, regardless of the underlying etiology, heart failure is always associated with both local and systemic activation of inflammatory signaling cascades. Cardiac macrophages are nodal regulators of inflammation. Resident macrophages mostly attenuate cardiac injury by secreting cytoprotective factors (cytokines, chemokines, and growth factors), scavenging damaged cells or mitochondrial debris, and regulating cardiac conduction, angiogenesis, lymphangiogenesis, and fibrosis. In contrast, excessive recruitment of monocyte-derived inflammatory macrophages largely contributes to the transition to heart failure. The current review examines the ambivalent role of inflammation (mainly TNFα-related) and cardiac macrophages (Mφ) in pathophysiologies from non-infarction origin, focusing on the protective signaling processes. Our objective is to illustrate how harnessing this knowledge could pave the way for innovative therapeutics in patients with heart failure.
Collapse
Affiliation(s)
| | | | | | - Catherine Pavoine
- INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, UMR_S1166, F-75013 Paris, France; (S.B.); (S.N.); (E.B.)
| |
Collapse
|
30
|
Bailey LR, Davis J. If these myocytes could talk, they would speak the language of metabolites. J Clin Invest 2022; 132:156296. [PMID: 35040436 PMCID: PMC8759775 DOI: 10.1172/jci156296] [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] [Indexed: 11/25/2022] Open
Abstract
Cardiac wound healing following ischemic injury requires a well-described spatiotemporal progression of events involving multiple cell types and cell-cell interactions. While cellular crosstalk among immune cell, endothelial cell, and fibroblast populations is known to regulate these progressive phases, the role of cardiac myocytes in controlling the wound-healing program is unclear. In this issue of the JCI, Li et al. describe a mechanism of cellular crosstalk between cardiac myocytes and fibroblasts that disrupts nonmyocyte cell function and worsens wound healing outcomes following myocardial infarction (MI). This tour de force study used an arsenal of multidisciplinary approaches to identify a central role for the ectonucleotidase ENPP1 in this process. These findings have clear therapeutic implications, as the authors identified a small molecular inhibitor of ENPP1 that improved post-MI outcomes in mice. These exciting data provide impactful mechanistic information that advance the field’s understanding of cardiac repair and remodeling.
Collapse
Affiliation(s)
- Logan Rj Bailey
- Molecular and Cellular Biology Graduate Program.,Medical Scientist Training Program
| | - Jennifer Davis
- Department of Bioengineering.,Department of Lab Medicine and Pathology.,Institute for Stem Cell and Regenerative Medicine, and.,Center for Cardiovascular Biology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
31
|
Sorokina AG, Orlova YA, Grigorieva OA, Novoseletskaya ES, Basalova NA, Alexandrushkina NA, Vigovsky MA, Kirillova KI, Balatsky AV, Samokhodskaya LM, Danilova NV, Dyachkova UD, Fedotov DА, Akopyan AA, Kakotkin VV, Asratyan DA, Akopyan ZA, Efimenko AY. Creation of a collection of different biological sample types from elderly patients to study the relationship of clinical, systemic, tissue and cellular biomarkers of accumulation of senescent cells during aging. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2022. [DOI: 10.15829/1728-8800-2021-3051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
With aging, tissue homeostasis and their effective recovery after damage is violated. It has been shown that this may be due to the excessive accumulation of senescent (SC) cells in various tissues, which leads to the activation of chronic sterile inflammation, tissue dysfunction and, as a result, to the development of age-related diseases. To assess the contribution of SC cells to human body aging and pathogenesis of such diseases, relevant biomarkers are studied. For successful translation into clinical practice of approaches aimed at regulating the SC cell content in various tissues, it is necessary to study the relationship between the established clinical biomarkers of aging and age-related diseases, systemic aging parameters, and SC biomarkers at the tissue and cellular levels.Aim. To develop and describe action algorithms for creating a biobank of samples obtained from patients aged >65 years in order to study biomarkers of SC cell accumulation.Material and methods. To collect samples, an interaction system was built between several research, clinical and infrastructure departments of a multidisciplinary medical center. At the stage of preanalytical training, regulatory legal acts were developed, including informed consent for patients, as well as protocols for each stage of the study.Results. A roadmap was formed with action algorithms for all participants in the study, as well as with a convenient and accessible system of annotations and storage of biological samples. To date, the collection includes biological samples of 7 different types (peripheral blood serum, formalin-fixed tissue samples and formalin fixed paraffin embedded tissue specimens, samples of different cells isolated from peripheral blood, skin and adipose tissue, samples of deoxyribonucleic and ribonucleic acids, cell secretome conditioned media) obtained from 82 patients. We accumulated relevant anamnestic, clinical and laboratory data, as well as the results of experimental studies to assess the SC cell biomarkers. Using the collection, the relationship between clinical, tissue and cellular biomarkers of SC cell accumulation was studied.Conclusion. The creation of a collection of biological samples at the molecular, cellular, tissue and organism levels from one patient provides great opportunities for research in the field of personalized medicine and the study of age-related disease pathogenesis.
Collapse
Affiliation(s)
- A. G. Sorokina
- Medical Research and Education Center of the Lomonosov Moscow State University
| | - Ya. A. Orlova
- Medical Research and Education Center of the Lomonosov Moscow State University; Lomonosov Moscow State University
| | - O. A. Grigorieva
- Medical Research and Education Center of the Lomonosov Moscow State University
| | - E. S. Novoseletskaya
- Medical Research and Education Center of the Lomonosov Moscow State University; Lomonosov Moscow State University
| | - N. A. Basalova
- Medical Research and Education Center of the Lomonosov Moscow State University; Lomonosov Moscow State University
| | - N. A. Alexandrushkina
- Medical Research and Education Center of the Lomonosov Moscow State University; Lomonosov Moscow State University
| | - M. A. Vigovsky
- Medical Research and Education Center of the Lomonosov Moscow State University; Lomonosov Moscow State University
| | - K. I. Kirillova
- Medical Research and Education Center of the Lomonosov Moscow State University
| | - A. V. Balatsky
- Medical Research and Education Center of the Lomonosov Moscow State University
| | - L. M. Samokhodskaya
- Medical Research and Education Center of the Lomonosov Moscow State University; Lomonosov Moscow State University
| | - N. V. Danilova
- Medical Research and Education Center of the Lomonosov Moscow State University; Lomonosov Moscow State University
| | | | | | - A. A. Akopyan
- Medical Research and Education Center of the Lomonosov Moscow State University
| | | | - D. A. Asratyan
- Medical Research and Education Center of the Lomonosov Moscow State University
| | - Zh. A. Akopyan
- Medical Research and Education Center of the Lomonosov Moscow State University; Lomonosov Moscow State University
| | - A. Yu. Efimenko
- Medical Research and Education Center of the Lomonosov Moscow State University; Lomonosov Moscow State University
| |
Collapse
|
32
|
Waleczek FJG, Sansonetti M, Xiao K, Jung M, Mitzka S, Dendorfer A, Weber N, Perbellini F, Thum T. Chemical and mechanical activation of resident cardiac macrophages in the living myocardial slice ex vivo model. Basic Res Cardiol 2022; 117:63. [PMID: 36449104 PMCID: PMC9712328 DOI: 10.1007/s00395-022-00971-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 11/16/2022] [Accepted: 11/16/2022] [Indexed: 12/02/2022]
Abstract
Resident cardiac macrophages (rcMACs) are among the most abundant immune cells in the heart. Plasticity and activation are hallmarks of rcMACs in response to changes in the microenvironment, which is essential for in vitro experimentation. The in vivo investigation is confounded by the infiltration of other cells hindering direct studies of rcMACs. As a tool to investigate rcMACs, we applied the ex vivo model of living myocardial slices (LMS). LMS are ultrathin ex vivo multicellular cardiac preparations in which the circulatory network is interrupted. The absence of infiltration in this model enables the investigation of the rcMACs response to immunomodulatory and mechanical stimulations. Such conditions were generated by applying interferon-gamma (IFN-γ) or interleukine-4 (IL-4) and altering the preload of cultured LMS, respectively. The immunomodulatory stimulation of the LMS induced alterations of the gene expression pattern without affecting tissue contractility. Following 24 h culture, low input RNA sequencing of rcMACs isolated from LMS was used for gene ontology analysis. Reducing the tissue stretch (unloading) of LMS altered the gene ontology clusters of isolated rcMACs with intermediate semantic similarity to IFN-γ triggered reaction. Through the overlap of genes affected by IFN-γ and unloading, we identified Allograft inflammatory factor 1 (AIF-1) as a potential marker gene for inflammation of rcMACs as significantly altered in whole immunomodulated LMS. MicroRNAs associated with the transcriptomic changes of rcMACs in unloaded LMS were identified in silico. Here, we demonstrate the approach of LMS to understand load-triggered cardiac inflammation and, thus, identify potential translationally important therapeutic targets.
Collapse
Affiliation(s)
- F. J. G. Waleczek
- grid.10423.340000 0000 9529 9877Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany
| | - M. Sansonetti
- grid.10423.340000 0000 9529 9877Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany
| | - K. Xiao
- grid.10423.340000 0000 9529 9877Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany ,grid.4561.60000 0000 9261 3939Fraunhofer Institute ITEM, Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany
| | - M. Jung
- grid.10423.340000 0000 9529 9877Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany
| | - S. Mitzka
- grid.10423.340000 0000 9529 9877Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany ,grid.4561.60000 0000 9261 3939Fraunhofer Institute ITEM, Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany
| | - A. Dendorfer
- grid.5252.00000 0004 1936 973XWalter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University München, Marchioninistraße 27, 81377 Munich, Germany
| | - N. Weber
- grid.10423.340000 0000 9529 9877Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany
| | - F. Perbellini
- grid.10423.340000 0000 9529 9877Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany
| | - T. Thum
- grid.10423.340000 0000 9529 9877Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany ,grid.4561.60000 0000 9261 3939Fraunhofer Institute ITEM, Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany
| |
Collapse
|
33
|
[SEMERGEN positioning on approaching chronic heart failure in primary care]. Semergen 2021; 48:106-123. [PMID: 34924298 DOI: 10.1016/j.semerg.2021.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/24/2021] [Accepted: 10/27/2021] [Indexed: 12/11/2022]
Abstract
Heart failure (HF) is a public health problem that generates a large healthcare burden both in hospitals and in Primary Care (PC). The publication of numerous studies about HF in recent years has led to a paradigm shift in the approach to this syndrome, in which the work of PC teams is gaining greater prominence. The recent guidelines published by the European Society of Cardiology have fundamentally introduced changes in the management of patients with HF. The new proposed strategy, with drugs that reduce hospitalizations and slow the progression of the disease, should now be a priority for all professionals involved. This position document analyzes a proposal for an approach based on multidisciplinary teams with the leadership of family doctors, key to providing quality care throughout the entire process of the disease, from its prevention to the end of the life.
Collapse
|
34
|
Yeh CH, Chou YJ, Chu TK, Tsai TF. Rejuvenating the Aging Heart by Enhancing the Expression of the Cisd2 Prolongevity Gene. Int J Mol Sci 2021; 22:ijms222111487. [PMID: 34768917 PMCID: PMC8583758 DOI: 10.3390/ijms222111487] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 02/07/2023] Open
Abstract
Aging is the major risk factor for cardiovascular disease, which is the leading cause of mortality worldwide among aging populations. Cisd2 is a prolongevity gene that mediates lifespan in mammals. Previously, our investigations revealed that a persistently high level of Cisd2 expression in mice is able to prevent age-associated cardiac dysfunction. This study was designed to apply a genetic approach that induces cardiac-specific Cisd2 overexpression (Cisd2 icOE) at a late-life stage, namely a time point immediately preceding the onset of old age, and evaluate the translational potential of this approach. Several discoveries are pinpointed. Firstly, Cisd2 is downregulated in the aging heart. This decrease in Cisd2 leads to cardiac dysfunction and impairs electromechanical performance. Intriguingly, Cisd2 icOE prevents an exacerbation of age-associated electromechanical dysfunction. Secondly, Cisd2 icOE ameliorates cardiac fibrosis and improves the integrity of the intercalated discs, thereby reversing various structural abnormalities. Finally, Cisd2 icOE reverses the transcriptomic profile of the aging heart, changing it from an older-age pattern to a younger pattern. Intriguingly, Cisd2 icOE modulates a number of aging-related pathways, namely the sirtuin signaling, autophagy, and senescence pathways, to bring about rejuvenation of the heart as it enters old age. Our findings highlight Cisd2 as a novel molecular target for developing therapies targeting cardiac aging.
Collapse
Affiliation(s)
- Chi-Hsiao Yeh
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
| | - Yi-Ju Chou
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Zhunan, Miaoli 350, Taiwan;
| | - Ting-Kuan Chu
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
| | - Ting-Fen Tsai
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Zhunan, Miaoli 350, Taiwan;
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
- Aging and Health Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Correspondence: ; Tel.: +886-2-28267293
| |
Collapse
|
35
|
Bauersachs J, López-Andrés N. Mineralocorticoid receptor in cardiovascular diseases-Clinical trials and mechanistic insights. Br J Pharmacol 2021; 179:3119-3134. [PMID: 34643952 DOI: 10.1111/bph.15708] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/07/2021] [Accepted: 09/27/2021] [Indexed: 12/19/2022] Open
Abstract
Aldosterone binds to the mineralocorticoid receptor (NR3C2), a transcription factor of the nuclear receptor family, present in the kidney and in various other non-epithelial cells including the heart and the vasculature. Indeed, extra-renal pathophysiological effects of this hormone have been characterized, extending its actions to the cardiovascular system. A growing body of clinical and pre-clinical evidence suggests that mineralocorticoid receptor overactivation plays an important pathophysiological role in cardiovascular remodelling by promoting cardiac hypertrophy, fibrosis, arterial stiffness and in inflammation and oxidative stress. The following review article outlines the role of mineralocorticoid receptor in cardiovascular disease with a focus on myocardial remodelling and heart failure (HF) including clinical trials as well as cellular and animal studies.
Collapse
Affiliation(s)
- Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Natalia López-Andrés
- Cardiovascular Translational Research. Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| |
Collapse
|
36
|
Taneja A, Ravi V, Hong JY, Lin H, Sundaresan NR. Emerging roles of Sirtuin 2 in cardiovascular diseases. FASEB J 2021; 35:e21841. [PMID: 34582046 DOI: 10.1096/fj.202100490r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 07/07/2021] [Accepted: 07/23/2021] [Indexed: 11/11/2022]
Abstract
Sirtuins are a family of NAD+ -dependent deacetylases implicated in a wide variety of age-associated pathologies, including cardiovascular disorders. Among the seven mammalian sirtuins, SIRT2 modulates various cellular processes through the deacetylation or deacylation of their target proteins. Notably, the levels of SIRT2 in the heart decline with age and other pathological conditions, leading to cardiovascular dysfunction. In the present review, we discuss the emerging roles of SIRT2 in cardiovascular dysfunction and heart failure associated with factors like age, hypertension, oxidative stress, and diabetes. We also discuss the potential of using inhibitors to study the unexplored role of SIRT2 in the heart. While SIRT2 undoubtedly plays a crucial role in the cardiovascular system, its functions are only beginning to be understood, making it an attractive candidate for further research in the field.
Collapse
Affiliation(s)
- Arushi Taneja
- Department of Microbiology and Cell Biology, Cardiovascular and Muscle Research Laboratory, Indian Institute of Science, Bengaluru, India
| | - Venkatraman Ravi
- Department of Microbiology and Cell Biology, Cardiovascular and Muscle Research Laboratory, Indian Institute of Science, Bengaluru, India
| | - Jun Young Hong
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA
| | - Hening Lin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA.,Howard Hughes Medical Institute, Cornell University, Ithaca, NY, USA
| | - Nagalingam Ravi Sundaresan
- Department of Microbiology and Cell Biology, Cardiovascular and Muscle Research Laboratory, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
37
|
Tryptophan: From Diet to Cardiovascular Diseases. Int J Mol Sci 2021; 22:ijms22189904. [PMID: 34576067 PMCID: PMC8472285 DOI: 10.3390/ijms22189904] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/02/2021] [Accepted: 09/11/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular disease (CVD) is one of the major causes of mortality worldwide. Inflammation is the underlying common mechanism involved in CVD. It has been recently related to amino acid metabolism, which acts as a critical regulator of innate and adaptive immune responses. Among different metabolites that have emerged as important regulators of immune and inflammatory responses, tryptophan (Trp) metabolites have been shown to play a pivotal role in CVD. Here, we provide an overview of the fundamental aspects of Trp metabolism and the interplay between the dysregulation of the main actors involved in Trp metabolism such as indoleamine 2, 3-dioxygenase 1 (IDO) and CVD, including atherosclerosis and myocardial infarction. IDO has a prominent and complex role. Its activity, impacting on several biological pathways, complicates our understanding of its function, particularly in CVD, where it is still under debate. The discrepancy of the observed IDO effects could be potentially explained by its specific cell and tissue contribution, encouraging further investigations regarding the role of this enzyme. Thus, improving our understanding of the function of Trp as well as its derived metabolites will help to move one step closer towards tailored therapies aiming to treat CVD.
Collapse
|
38
|
Mesquita T, Lin Y, Ibrahim A. Chronic low-grade inflammation in heart failure with preserved ejection fraction. Aging Cell 2021; 20:e13453. [PMID: 34382743 PMCID: PMC8441359 DOI: 10.1111/acel.13453] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/21/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023] Open
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is currently the predominant form of HF with a dramatic increase in risk with age. Low-grade inflammation, as occurs with aging (termed "inflammaging"), is a common feature of HFpEF pathology. Suppression of proinflammatory pathways has been associated with attenuated HFpEF disease severity and better outcomes. From this perspective, inflammasome signaling plays a central role in mediating chronic inflammation and cardiovascular disease progression. However, the causal link between the inflammasome-immune signaling axis on the age-dependent progression of HFpEF remains conjectural. In this review, we summarize the current understanding of the role of inflammatory pathways in age-dependent cardiac function decline. We will also evaluate recent advances and evidence regarding the inflammatory pathway in the pathophysiology of HFpEF, with special attention to inflammasome signaling.
Collapse
Affiliation(s)
- Thassio Mesquita
- Cedars‐Sinai Medical CenterSmidt Heart InstituteLos AngelesCAUSA
| | - Yen‐Nien Lin
- Cedars‐Sinai Medical CenterSmidt Heart InstituteLos AngelesCAUSA
- Division of Cardiovascular MedicineDepartment of MedicineChina Medical University and HospitalTaichungTaiwan
| | - Ahmed Ibrahim
- Cedars‐Sinai Medical CenterSmidt Heart InstituteLos AngelesCAUSA
| |
Collapse
|
39
|
Wagner JUG, Dimmeler S. The endothelial niche in heart failure: from development to regeneration. Eur Heart J 2021; 42:4277-4279. [PMID: 34392349 DOI: 10.1093/eurheartj/ehab304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Julian U G Wagner
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Frankfurt, Germany.,Cardio-Pulmonary Institute (CPI), Frankfurt, Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Frankfurt, Germany.,Cardio-Pulmonary Institute (CPI), Frankfurt, Germany
| |
Collapse
|
40
|
Abstract
Macrophages are essential components of the immune system and play a role in the normal functioning of the cardiovascular system. Depending on their origin and phenotype, cardiac macrophages perform various functions. In a steady-state, these cells play a beneficial role in maintaining cardiac homeostasis by defending the body from pathogens and eliminating apoptotic cells, participating in electrical conduction, vessel patrolling, and arterial tone regulation. However, macrophages also take part in adverse cardiac remodeling that could lead to the development and progression of heart failure (HF) in such HF comorbidities as hypertension, obesity, diabetes, and myocardial infarction. Nevertheless, studies on detailed mechanisms of cardiac macrophage function are still in progress, and could enable potential therapeutic applications of these cells. This review aims to present the latest reports on the origin, heterogeneity, and functions of cardiac macrophages in the healthy heart and in cardiovascular diseases leading to HF. The potential therapeutic use of macrophages is also briefly discussed.
Collapse
|
41
|
Shen YH, Abe JI. Nrg1β as a Proangiogenesis Therapy: How Is Nrg1β Therapy Unique From Other Angiogenesis Therapies? Arterioscler Thromb Vasc Biol 2021; 41:2315-2317. [PMID: 34162231 DOI: 10.1161/atvbaha.121.316513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Ying H Shen
- Division of Cardiothoracic Surgery, Baylor College of Medicine, Houston, TX (Y.H.S.)
| | - Jun-Ichi Abe
- Division of Internal Medicine, Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston (J.I.A.)
| |
Collapse
|
42
|
Application of genetic cell-lineage tracing technology to study cardiovascular diseases. J Mol Cell Cardiol 2021; 156:57-68. [PMID: 33745891 DOI: 10.1016/j.yjmcc.2021.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/03/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular diseases are leading causes that threaten people's life. To investigate cells that are involved in disease development and tissue repair, various technologies have been introduced. Among these technologies, lineage tracing is a powerful tool to track the fate of cells in vivo, providing deep insights into cellular behavior and plasticity. In cardiac diseases, newly formed cardiomyocytes and endothelial cells are found from proliferation of local cells, while fibroblasts and macrophages are originated from diverse cell sources. Similarly, in response to vascular injury, various sources of cells including media smooth muscle cells, endothelium, resident progenitors and bone marrow cells are involved in lesion formation and/or vessel regeneration. In summary, current review summarizes the development of lineage tracing techniques and their utilizations in investigating roles of different cell types in cardiovascular diseases.
Collapse
|
43
|
Segovia-Roldan M, Diez ER, Pueyo E. Melatonin to Rescue the Aged Heart: Antiarrhythmic and Antioxidant Benefits. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8876792. [PMID: 33791076 PMCID: PMC7984894 DOI: 10.1155/2021/8876792] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 01/16/2021] [Accepted: 01/23/2021] [Indexed: 12/19/2022]
Abstract
Aging comes with gradual loss of functions that increase the vulnerability to disease, senescence, and death. The mechanisms underlying these processes are linked to a prolonged imbalance between damage and repair. Damaging mechanisms include oxidative stress, mitochondrial dysfunction, chronodisruption, inflammation, and telomere attrition, as well as genetic and epigenetic alterations. Several endogenous tissue repairing mechanisms also decrease. These alterations associated with aging affect the entire organism. The most devastating manifestations involve the cardiovascular system and may lead to lethal cardiac arrhythmias. Together with structural remodeling, electrophysiological and intercellular communication alterations during aging predispose to arrhythmic events. Despite the knowledge on repairing mechanisms in the cardiovascular system, effective antiaging strategies able to reduce the risk of arrhythmias are still missing. Melatonin is a promising therapeutic candidate due to its pleiotropic actions. This indoleamine regulates chronobiology and endocrine physiology. Of relevance, melatonin is an antiaging, antioxidant, antiapoptotic, antiarrhythmic, immunomodulatory, and antiproliferative molecule. This review focuses on the protective effects of melatonin on age-induced cardiac functional and structural alterations, potentially becoming a new fountain of youth for the heart.
Collapse
Affiliation(s)
- Margarita Segovia-Roldan
- Biomedical Signal Interpretation and Computational Simulation (BSICoS), I3A, Universidad de Zaragoza, IIS Aragón and CIBER-BBN, Spain
| | | | - Esther Pueyo
- Biomedical Signal Interpretation and Computational Simulation (BSICoS), I3A, Universidad de Zaragoza, IIS Aragón and CIBER-BBN, Spain
| |
Collapse
|
44
|
Cai Y, Liu H, Song E, Wang L, Xu J, He Y, Zhang D, Zhang L, Cheng KKY, Jin L, Wu M, Liu S, Qi D, Zhang L, Lopaschuk GD, Wang S, Xu A, Xia Z. Deficiency of telomere-associated repressor activator protein 1 precipitates cardiac aging in mice via p53/PPARα signaling. Theranostics 2021; 11:4710-4727. [PMID: 33754023 PMCID: PMC7978321 DOI: 10.7150/thno.51739] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 02/15/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Telomere shortening and dysfunction may cause metabolic disorders, tissue damage and age-dependent pathologies. However, little is known about the association of telomere-associated protein Rap1 with mitochondrial energy metabolism and cardiac aging. Methods: Echocardiography was performed to detect cardiac structure and function in Rap1+/+ and Rap1-/- mice at different ages (3 months, 12 months and 20 months). Telomere length, DNA damage, cardiac senescence and cardiomyocyte size were analyzed using the real-time PCR, Western blotting, senescence associated β-galactosidase assay and wheat germ agglutinin staining, respectively. Western blotting was also used to determine the level of cardiac fatty acid metabolism related key enzymes in mouse and human myocardium. Chromatin immunoprecipitation assay was used to verify the direct link between p53 and PPARα. The p53 inhibitor, Pifithrin-α and PPARα activator WY14643 were utilized to identify the effects of Rap1/p53/PPARα signaling pathway. Results: Telomere was shortened concomitant with extensive DNA damage in aged Rap1-/- mouse hearts, evidenced by reduced T/S ratios and increased nuclear γH2AX. Meanwhile, the aging-associated phenotypes were pronounced as reflected by altered mitochondrial ultrastructure, enhanced senescence, cardiac hypertrophy and dysfunction. Mechanistically, acetylated p53 and nuclear p53 was enhanced in the Rap1-/- mouse hearts, concomitant with reduced PPARα. Importantly, p53 directly binds to the promoter of PPARα in mouse hearts and suppresses the transcription of PPARα. In addition, aged Rap1-/- mice exhibited reduced cardiac fatty acid metabolism. Pifithrin-α alleviated cardiac aging and enhanced fatty acid metabolism in the aged Rap1-/- mice. Activating PPARα with WY14643 in primarily cultured Rap1-/- cardiomyocytes restored maximal oxygen consumption rates. Reduced Rap1 expression and impaired p53/PPARα signaling also presented in aged human myocardium. Conclusion: In summary, Rap1 may link telomere biology to fatty acid metabolism and aging-related cardiac pathologies via modulating the p53/PPARα signaling pathway, which could represent a therapeutic target in preventing/attenuating cardiac aging.
Collapse
|
45
|
Nicin L, Abplanalp WT, Schänzer A, Sprengel A, John D, Mellentin H, Tombor L, Keuper M, Ullrich E, Klingel K, Dettmeyer RB, Hoffmann J, Akintuerk H, Jux C, Schranz D, Zeiher AM, Rupp S, Dimmeler S. Single Nuclei Sequencing Reveals Novel Insights Into the Regulation of Cellular Signatures in Children With Dilated Cardiomyopathy. Circulation 2021; 143:1704-1719. [PMID: 33618539 DOI: 10.1161/circulationaha.120.051391] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a leading cause of death in children with heart failure. The outcome of pediatric heart failure treatment is inconsistent, and large cohort studies are lacking. Progress may be achieved through personalized therapy that takes age- and disease-related pathophysiology, pathology, and molecular fingerprints into account. We present single nuclei RNA sequencing from pediatric patients with DCM as the next step in identifying cellular signatures. METHODS We performed single nuclei RNA sequencing with heart tissues from 6 children with DCM with an age of 0.5, 0.75, 5, 6, 12, and 13 years. Unsupervised clustering of 18 211 nuclei led to the identification of 14 distinct clusters with 6 major cell types. RESULTS The number of nuclei in fibroblast clusters increased with age in patients with DCM, a finding that was confirmed by histological analysis and was consistent with an age-related increase in cardiac fibrosis quantified by cardiac magnetic resonance imaging. Fibroblasts of patients with DCM >6 years of age showed a profoundly altered gene expression pattern with enrichment of genes encoding fibrillary collagens, modulation of proteoglycans, switch in thrombospondin isoforms, and signatures of fibroblast activation. In addition, a population of cardiomyocytes with a high proregenerative profile was identified in infant patients with DCM but was absent in children >6 years of age. This cluster showed high expression of cell cycle activators such as cyclin D family members, increased glycolytic metabolism and antioxidative genes, and alterations in ß-adrenergic signaling genes. CONCLUSIONS Novel insights into the cellular transcriptomes of hearts from pediatric patients with DCM provide remarkable age-dependent changes in the expression patterns of fibroblast and cardiomyocyte genes with less fibrotic but enriched proregenerative signatures in infants.
Collapse
Affiliation(s)
- Luka Nicin
- Institute for Cardiovascular Regeneration (L.N., W.T.A., D.J., H.M., L.T., S.D.), Goethe University, Germany.,German Center for Cardiovascular Research, Frankfurt, Germany (L.N., W.T.A., S.D.).,Cardio-Pulmonary Institute, Frankfurt, Germany (L.N., W.T.A., S.D.)
| | - Wesley T Abplanalp
- Institute for Cardiovascular Regeneration (L.N., W.T.A., D.J., H.M., L.T., S.D.), Goethe University, Germany.,German Center for Cardiovascular Research, Frankfurt, Germany (L.N., W.T.A., S.D.).,Cardio-Pulmonary Institute, Frankfurt, Germany (L.N., W.T.A., S.D.)
| | - Anne Schänzer
- Institute of Neuropathology (A.S., M.K.), University Hospital Giessen, Justus Liebig Universität, Germany
| | - Anke Sprengel
- Pediatric Heart Center, Department of Pediatric Cardiac Surgery (A.S., H.A.), University Hospital Giessen, Justus Liebig Universität, Germany
| | - David John
- Institute for Cardiovascular Regeneration (L.N., W.T.A., D.J., H.M., L.T., S.D.), Goethe University, Germany
| | - Hannah Mellentin
- Institute for Cardiovascular Regeneration (L.N., W.T.A., D.J., H.M., L.T., S.D.), Goethe University, Germany
| | - Lukas Tombor
- Institute for Cardiovascular Regeneration (L.N., W.T.A., D.J., H.M., L.T., S.D.), Goethe University, Germany
| | - Matthias Keuper
- Institute of Neuropathology (A.S., M.K.), University Hospital Giessen, Justus Liebig Universität, Germany
| | - Evelyn Ullrich
- Experimental Immunology, Division of Pediatric Stem Cell Transplantation and Immunology, Children and Adolescents Medicine, University Hospital Frankfurt (E.U.), Goethe University, Germany.,Frankfurt Cancer Institute (E.U.), Goethe University, Germany
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tuebingen, Germany (K.K.)
| | | | - Jedrzej Hoffmann
- Internal Medicine Clinic III, Department of Cardiology (J.H., A.M.Z.), Goethe University, Germany
| | - Hakan Akintuerk
- Pediatric Heart Center, Department of Pediatric Cardiac Surgery (A.S., H.A.), University Hospital Giessen, Justus Liebig Universität, Germany
| | - Christian Jux
- Department of Pediatric Cardiology and Congenital Heart Disease (C.J., D.S., S.R.), University Hospital Giessen, Justus Liebig Universität, Germany
| | - Dietmar Schranz
- Department of Pediatric Cardiology and Congenital Heart Disease (C.J., D.S., S.R.), University Hospital Giessen, Justus Liebig Universität, Germany
| | - Andreas M Zeiher
- Internal Medicine Clinic III, Department of Cardiology (J.H., A.M.Z.), Goethe University, Germany
| | - Stefan Rupp
- Department of Pediatric Cardiology and Congenital Heart Disease (C.J., D.S., S.R.), University Hospital Giessen, Justus Liebig Universität, Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration (L.N., W.T.A., D.J., H.M., L.T., S.D.), Goethe University, Germany.,German Center for Cardiovascular Research, Frankfurt, Germany (L.N., W.T.A., S.D.).,Cardio-Pulmonary Institute, Frankfurt, Germany (L.N., W.T.A., S.D.)
| |
Collapse
|
46
|
Ramanujam D, Schön AP, Beck C, Vaccarello P, Felician G, Dueck A, Esfandyari D, Meister G, Meitinger T, Schulz C, Engelhardt S. MicroRNA-21-Dependent Macrophage-to-Fibroblast Signaling Determines the Cardiac Response to Pressure Overload. Circulation 2021; 143:1513-1525. [PMID: 33550817 PMCID: PMC8032214 DOI: 10.1161/circulationaha.120.050682] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Supplemental Digital Content is available in the text. Background: Cardiac macrophages (cMPs) are increasingly recognized as important regulators of myocardial homeostasis and disease, yet the role of noncoding RNA in these cells is largely unknown. Small RNA sequencing of the entire miRNomes of the major cardiac cell fractions revealed microRNA-21 (miR-21) as the single highest expressed microRNA in cMPs, both in health and disease (25% and 43% of all microRNA reads, respectively). MiR-21 has been previously reported as a key microRNA driving tissue fibrosis. Here, we aimed to determine the function of macrophage miR-21 on myocardial homeostasis and disease-associated remodeling. Methods: Macrophage-specific ablation of miR-21 in mice driven by Cx3cr1-Cre was used to determine the function of miR-21 in this cell type. As a disease model, mice were subjected to pressure overload for 6 and 28 days. Cardiac function was assessed in vivo by echocardiography, followed by histological analyses and single-cell sequencing. Cocultures of macrophages and cardiac fibroblasts were used to study macrophage-to-fibroblast signaling. Results: Mice with macrophage-specific genetic deletion of miR-21 were protected from interstitial fibrosis and cardiac dysfunction when subjected to pressure overload of the left ventricle. Single-cell sequencing of pressure-overloaded hearts from these mice revealed that miR-21 in macrophages is essential for their polarization toward a M1-like phenotype. Systematic quantification of intercellular communication mediated by ligand-receptor interactions across all cell types revealed that miR-21 primarily determined macrophage-fibroblast communication, promoting the transition from quiescent fibroblasts to myofibroblasts. Polarization of isolated macrophages in vitro toward a proinflammatory (M1-like) phenotype activated myofibroblast transdifferentiation of cardiac fibroblasts in a paracrine manner and was dependent on miR-21 in cMPs. Conclusions: Our data indicate a critical role of cMPs in pressure overload–induced cardiac fibrosis and dysfunction and reveal macrophage miR-21 as a key molecule for the profibrotic role of cMPs.
Collapse
Affiliation(s)
- Deepak Ramanujam
- Institut für Pharmakologie und Toxikologie (D.R., A.P.S., C.B., P.V., G.F., A.D., D.E., S.E.), Technische Universität München (TUM), Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (D.R., A.P.S., C.B., G.F., A.D., D.E., T.M., C.S., S.E.)
| | - Anna Patricia Schön
- Institut für Pharmakologie und Toxikologie (D.R., A.P.S., C.B., P.V., G.F., A.D., D.E., S.E.), Technische Universität München (TUM), Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (D.R., A.P.S., C.B., G.F., A.D., D.E., T.M., C.S., S.E.)
| | - Christina Beck
- Institut für Pharmakologie und Toxikologie (D.R., A.P.S., C.B., P.V., G.F., A.D., D.E., S.E.), Technische Universität München (TUM), Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (D.R., A.P.S., C.B., G.F., A.D., D.E., T.M., C.S., S.E.)
| | - Paula Vaccarello
- Institut für Pharmakologie und Toxikologie (D.R., A.P.S., C.B., P.V., G.F., A.D., D.E., S.E.), Technische Universität München (TUM), Germany
| | - Giulia Felician
- Institut für Pharmakologie und Toxikologie (D.R., A.P.S., C.B., P.V., G.F., A.D., D.E., S.E.), Technische Universität München (TUM), Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (D.R., A.P.S., C.B., G.F., A.D., D.E., T.M., C.S., S.E.)
| | - Anne Dueck
- Institut für Pharmakologie und Toxikologie (D.R., A.P.S., C.B., P.V., G.F., A.D., D.E., S.E.), Technische Universität München (TUM), Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (D.R., A.P.S., C.B., G.F., A.D., D.E., T.M., C.S., S.E.)
| | - Dena Esfandyari
- Institut für Pharmakologie und Toxikologie (D.R., A.P.S., C.B., P.V., G.F., A.D., D.E., S.E.), Technische Universität München (TUM), Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (D.R., A.P.S., C.B., G.F., A.D., D.E., T.M., C.S., S.E.)
| | - Gunter Meister
- Biochemistry Center Regensburg (BZR), Laboratory for RNA Biology, University of Regensburg, Germany (G.M.)
| | - Thomas Meitinger
- Institute of Human Genetics (T.M.), Technische Universität München (TUM), Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (D.R., A.P.S., C.B., G.F., A.D., D.E., T.M., C.S., S.E.).,Institut für Humangenetik, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany (T.M.)
| | - Christian Schulz
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (D.R., A.P.S., C.B., G.F., A.D., D.E., T.M., C.S., S.E.).,Medizinische Klinik und Poliklinik I, LMU Klinikum, Ludwig-Maximilians-Universität München, Germany (C.S.)
| | - Stefan Engelhardt
- Institut für Pharmakologie und Toxikologie (D.R., A.P.S., C.B., P.V., G.F., A.D., D.E., S.E.), Technische Universität München (TUM), Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (D.R., A.P.S., C.B., G.F., A.D., D.E., T.M., C.S., S.E.)
| |
Collapse
|
47
|
Garvin AM, Khokhar BS, Czubryt MP, Hale TM. RAS inhibition in resident fibroblast biology. Cell Signal 2020; 80:109903. [PMID: 33370581 DOI: 10.1016/j.cellsig.2020.109903] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023]
Abstract
Angiotensin II (Ang II) is a primary mediator of profibrotic signaling in the heart and more specifically, the cardiac fibroblast. Ang II-mediated cardiomyocyte hypertrophy in combination with cardiac fibroblast proliferation, activation, and extracellular matrix production compromise cardiac function and increase mortality in humans. Profibrotic actions of Ang II are mediated by increasing production of fibrogenic mediators (e.g. transforming growth factor beta, scleraxis, osteopontin, and periostin), recruitment of immune cells, and via increased reactive oxygen species generation. Drugs that inhibit Ang II production or action, collectively referred to as renin angiotensin system (RAS) inhibitors, are first line therapeutics for heart failure. Moreover, transient RAS inhibition has been found to persistently alter hypertensive cardiac fibroblast responses to injury providing a useful tool to identify novel therapeutic targets. This review summarizes the profibrotic actions of Ang II and the known impact of RAS inhibition on cardiac fibroblast phenotype and cardiac remodeling.
Collapse
Affiliation(s)
- Alexandra M Garvin
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Bilal S Khokhar
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Michael P Czubryt
- Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre and Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Taben M Hale
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, USA.
| |
Collapse
|
48
|
Xia W, Chen H, Chen D, Ye Y, Xie C, Hou M. PD-1 inhibitor inducing exosomal miR-34a-5p expression mediates the cross talk between cardiomyocyte and macrophage in immune checkpoint inhibitor-related cardiac dysfunction. J Immunother Cancer 2020; 8:jitc-2020-001293. [PMID: 33115945 PMCID: PMC7594538 DOI: 10.1136/jitc-2020-001293] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2020] [Indexed: 12/30/2022] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have been an important therapeutic advancement in the field of cancer medicine. Recent reports provided greater insights into the cardiovascular adverse events, which prohibited the use of ICIs. Cardiovascular adverse events occur in different forms, such as myocarditis and cardiomyopathy, myocardial fibrosis, heart failure and pericardial disease. Cardiac aging overlapped with the occurrence of some cardiac diseases. Exosomes mediate cell–cell cross talk in cardiac diseases by transferring a variety of biomolecules, including microRNAs (miRs). miR-34a-5p is a well-known miR associated with the cardiac senescence. This study aimed to investigate whether cardiovascular adverse effects of the programmed cell death 1 (PD-1) inhibitor, a widely used ICI, were related to exosomal-transferred miR-34a-5p in cardiac senescence in a mouse model. Methods and results The upregulation of miR-34a-5p in cardiomyocytes induced by exosomes derived from PD-1 inhibitor–treated macrophages, accompanied by cardiac senescence, caused cardiac injury in mouse hearts. miR-34a-5p was identified as an exosomal transfer RNA to induce cardiac senescence–related injury. Inhibiting miR-34a-5p in macrophages attenuated the exosomePD-1 inhibitor-induced pro-senescent effect in cardiomyocytes. TargetScan and luciferase assay showed that miR-34a-5p targeted the serine/threonine-protein phosphatase 1 regulatory subunit 10 (PNUTS) 3′-untranslated region. Conclusions Exosomes derived from PD-1 inhibitor–treated macrophages exerted a pro-senescent effect by modulating the miR-34a-5p/PNUTS signaling pathway. The findings might supply new targets to ameliorate cardiac injury in patients with cancer receiving PD-1 inhibitor treatment.
Collapse
Affiliation(s)
- Wenzheng Xia
- Neurosurgery, Wenzhou Medical University First Affiliated Hospital, Wenzhou, Zhejiang, China.,Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hanbin Chen
- Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Didi Chen
- Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yijia Ye
- Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Congying Xie
- Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Meng Hou
- Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
49
|
Jiang W, Liu M, Gu C, Ma H. The Pivotal Role of Mitsugumin 53 in Cardiovascular Diseases. Cardiovasc Toxicol 2020; 21:2-11. [PMID: 33006052 DOI: 10.1007/s12012-020-09609-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/23/2020] [Indexed: 12/28/2022]
Abstract
The MG53 (also known as TRIM72) is a conserved, muscle-specific tripartite motif family protein that is abundantly expressed in cardiac or skeletal muscle and present in circulation. Recently, the MG53 had been hypothesized to serve a dual role in the heart: involving in repairing cell membranes that protect myocardial function while acting as an E3 ligase to trigger insulin resistance and cardiovascular complications. This review discusses the roles of MG53 in cardiac physiological function with emphasis on MG53 protective function in the heart and its negative impact on the myocardium due to the continuous elevation of MG53. Besides, this work reviewed the significance of MG53 as a potential therapeutic in human cardiovascular diseases. Despite the expression of MG53 being rare in the human, thus exogenous MG53 can potentially be a new treatment for human cardiovascular diseases. Notably, the specific mechanism of MG53 in cardiovascular diseases remains elusive.
Collapse
Affiliation(s)
- Wenhua Jiang
- Institute of Medical Research, Northweastern Polytechnical University, Xi'an, 710072, People's Republic of China
| | - Manling Liu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Chunhu Gu
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China.
| | - Heng Ma
- Institute of Medical Research, Northweastern Polytechnical University, Xi'an, 710072, People's Republic of China.
| |
Collapse
|
50
|
Shimada BK, Yang Y, Zhu J, Wang S, Suen A, Kronstadt SM, Jeyaram A, Jay SM, Zou L, Chao W. Extracellular miR-146a-5p Induces Cardiac Innate Immune Response and Cardiomyocyte Dysfunction. Immunohorizons 2020; 4:561-572. [PMID: 32958516 PMCID: PMC7754174 DOI: 10.4049/immunohorizons.2000075] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 08/28/2020] [Indexed: 11/19/2022] Open
Abstract
Previous studies have demonstrated that transient myocardial ischemia leads to release of cellular nucleic acids such as RNA. Extracellular RNA reportedly plays a pivotal role in myocardial inflammation and ischemic injury in animals. RNA profiling has identified that numerous microRNA (miRNAs), such as ss-miR-146a-5p, are upregulated in plasma following myocardial ischemia, and certain uridine-rich miRNAs exhibit strong proinflammatory effects in immune cells via ssRNA-sensing mechanism. However, the effect of extracellular miRNAs on myocardial inflammation and cardiac cell function remains unknown. In this study, we treated adult mouse cardiomyocytes with miR-146a-5p loaded in extracellular vesicles and observed a dose- and TLR7-dependent production of CXCL-2, IL-6, and TNF-α. In vivo, a single dose of myocardial injection of miR-146a-5p induced both cytokine expression (CXCL2, IL-6, and TNF-α) and innate immune cell activation (CD45+ leukocytes, Ly6Cmid+ monocytes, Ly6G+ neutrophils), which was significantly attenuated in the hearts of TLR7 KO mice. We discovered that conditioned media from miR-146a-treated macrophages stimulated proinflammatory cytokine production in adult cardiomyocytes and significantly inhibited their sarcomere shortening. Finally, using an electric cell impedance-sensing assay, we found that the conditioned media from miR-146a-treated cardiac fibroblasts or cardiomyocytes impaired the barrier function of coronary artery endothelial cells. Taken together, these data demonstrate that extracellular miR-146a-5p activates multiple cardiac cells and induces myocardial inflammation and cardiomyocyte dysfunction via intercellular interaction and innate immune TLR7 nucleic acid sensing.
Collapse
Affiliation(s)
- Briana K Shimada
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201; and
| | - Yang Yang
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201; and
| | - Jing Zhu
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201; and
| | - Sheng Wang
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201; and
| | - Andrew Suen
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201; and
| | - Stephanie M Kronstadt
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20740
| | - Anjana Jeyaram
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20740
| | - Steven M Jay
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20740
| | - Lin Zou
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201; and
| | - Wei Chao
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201; and
| |
Collapse
|