1
|
Li J, Yao Y, Zhou J, Yang Z, Qiu C, Lu Y, Xie J, Liu J, Jiang T, Kou Y, Ge Z, Liang P, Qiu C, Shen L, Zhu Y, Gao C, Yu L. Epicardial transplantation of antioxidant polyurethane scaffold based human amniotic epithelial stem cell patch for myocardial infarction treatment. Nat Commun 2024; 15:9105. [PMID: 39438477 PMCID: PMC11496666 DOI: 10.1038/s41467-024-53531-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
Myocardial infarction (MI) is a leading cause of death globally. Stem cell therapy is considered a potential strategy for MI treatment. Transplantation of classic stem cells including embryonic, induced pluripotent and cardiac stem cells exhibited certain repairing effect on MI via supplementing cardiomyocytes, however, their clinical applications were blocked by problems of cell survival, differentiation, functional activity and also biosafety and ethical concerns. Here, we introduced human amniotic epithelial stem cells (hAESCs) featured with immunomodulatory activities, immune-privilege and biosafety, for constructing a stem cell cardiac patch based on porous antioxidant polyurethane (PUR), which demonstrated decent hAESCs compatibility. In rats, the administration of PUR-hAESC patch significantly reduced fibrosis and facilitated vascularization in myocardium after MI and consequently improved cardiac remodeling and function. Mechanistically, the patch provides a beneficial microenvironment for cardiac repair by facilitating a desirable immune response, paracrine modulation and limited oxidative milieu. Our findings may provide a potential therapeutic strategy for MI.
Collapse
Affiliation(s)
- Jinying Li
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine of Sir Run Run Shaw Hospital, Zhejiang University-Lishui Joint Innovation Center for Life and Health, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuejun Yao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Jiayi Zhou
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine of Sir Run Run Shaw Hospital, Zhejiang University-Lishui Joint Innovation Center for Life and Health, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhuoheng Yang
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine of Sir Run Run Shaw Hospital, Zhejiang University-Lishui Joint Innovation Center for Life and Health, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Chen Qiu
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine of Sir Run Run Shaw Hospital, Zhejiang University-Lishui Joint Innovation Center for Life and Health, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuwen Lu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Jieqi Xie
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Jia Liu
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine of Sir Run Run Shaw Hospital, Zhejiang University-Lishui Joint Innovation Center for Life and Health, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Tuoying Jiang
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine of Sir Run Run Shaw Hospital, Zhejiang University-Lishui Joint Innovation Center for Life and Health, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yaohui Kou
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine of Sir Run Run Shaw Hospital, Zhejiang University-Lishui Joint Innovation Center for Life and Health, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhen Ge
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310013, China
| | - Ping Liang
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Cong Qiu
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine of Sir Run Run Shaw Hospital, Zhejiang University-Lishui Joint Innovation Center for Life and Health, Zhejiang University, Hangzhou, 310058, China
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Liyin Shen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yang Zhu
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine of Sir Run Run Shaw Hospital, Zhejiang University-Lishui Joint Innovation Center for Life and Health, Zhejiang University, Hangzhou, 310058, China.
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China.
- State Key Laboratory of Transvascular Implantation Devices, Binjiang Institute of Zhejiang University, Hangzhou, 310053, China.
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China.
- Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China.
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China.
| | - Luyang Yu
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine of Sir Run Run Shaw Hospital, Zhejiang University-Lishui Joint Innovation Center for Life and Health, Zhejiang University, Hangzhou, 310058, China.
- College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
2
|
Lukovic D, Gyöngyösi M, Pavo IJ, Mester-Tonczar J, Einzinger P, Zlabinger K, Kastner N, Spannbauer A, Traxler D, Pavo N, Goliasch G, Pils D, Jakab A, Szankai Z, Michel-Behnke I, Zhang L, Devaux Y, Graf S, Beitzke D, Winkler J. Increased [ 18F]FDG uptake in the infarcted myocardial area displayed by combined PET/CMR correlates with snRNA-seq-detected inflammatory cell invasion. Basic Res Cardiol 2024; 119:807-829. [PMID: 38922408 PMCID: PMC11461641 DOI: 10.1007/s00395-024-01064-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
Combined [18F]FDG PET-cardiac MRI imaging (PET/CMR) is a useful tool to assess myocardial viability and cardiac function in patients with acute myocardial infarction (AMI). Here, we evaluated the prognostic value of PET/CMR in a porcine closed-chest reperfused AMI (rAMI) model. Late gadolinium enhancement by PET/CMR imaging displayed tracer uptake defect at the infarction site by 3 days after the rAMI in the majority of the animals (group Match, n = 28). Increased [18F]FDG uptake at the infarcted area (metabolism/contractility mismatch) with reduced tracer uptake in the remote viable myocardium (group Mismatch, n = 12) 3 days after rAMI was observed in the animals with larger infarct size and worse left ventricular ejection fraction (LVEF) (34 ± 8.7 vs 42.0 ± 5.2%), with lower LVEF also at the 1-month follow-up (35.8 ± 9.5 vs 43.0 ± 6.3%). Transcriptome analyses by bulk and single-nuclei RNA sequencing of the infarcted myocardium and border zones (n = 3 of each group, and 3 sham-operated controls) revealed a strong inflammatory response with infiltration of monocytes and macrophages in the infarcted and border areas in Mismatch animals. Our data indicate a high prognostic relevance of combined PET/MRI in the subacute phase of rAMI for subsequent impairment of heart function and underline the adverse effects of an excessive activation of the innate immune system in the initial phase after rAMI.
Collapse
Affiliation(s)
- Dominika Lukovic
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Mariann Gyöngyösi
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria.
| | - Imre J Pavo
- Division of Pediatric Cardiology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Julia Mester-Tonczar
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Patrick Einzinger
- Institute of Information Systems Engineering, Research Unit of Information and Software Engineering, Vienna University of Technology, 1040, Vienna, Austria
| | - Katrin Zlabinger
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Nina Kastner
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Andreas Spannbauer
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Denise Traxler
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Noemi Pavo
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Georg Goliasch
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Dietmar Pils
- Division of General Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Andras Jakab
- Center for MR-Research, University Children's Hospital Zurich, Zurich, Switzerland
| | | | - Ina Michel-Behnke
- Division of Pediatric Cardiology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Lu Zhang
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Senta Graf
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Dietrich Beitzke
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Johannes Winkler
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
3
|
Le L, Narula N, Zhou F, Smereka P, Ordner J, Theise N, Moore WH, Girvin F, Azour L, Moreira AL, Naidich DP, Ko JP. Diseases Involving the Lung Peribronchovascular Region: A CT Imaging Pathologic Classification. Chest 2024; 166:802-820. [PMID: 38909953 DOI: 10.1016/j.chest.2024.05.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 04/12/2024] [Accepted: 05/13/2024] [Indexed: 06/25/2024] Open
Abstract
TOPIC IMPORTANCE Chest CT imaging holds a major role in the diagnosis of lung diseases, many of which affect the peribronchovascular region. Identification and categorization of peribronchovascular abnormalities on CT imaging can assist in formulating a differential diagnosis and directing further diagnostic evaluation. REVIEW FINDINGS The peribronchovascular region of the lung encompasses the pulmonary arteries, airways, and lung interstitium. Understanding disease processes associated with structures of the peribronchovascular region and their appearances on CT imaging aids in prompt diagnosis. This article reviews current knowledge in anatomic and pathologic features of the lung interstitium composed of intercommunicating prelymphatic spaces, lymphatics, collagen bundles, lymph nodes, and bronchial arteries; diffuse lung diseases that present in a peribronchovascular distribution; and an approach to classifying diseases according to patterns of imaging presentations. Lung peribronchovascular diseases can appear on CT imaging as diffuse thickening, fibrosis, masses or masslike consolidation, ground-glass or air space consolidation, and cysts, acknowledging that some diseases may have multiple presentations. SUMMARY A category approach to peribronchovascular diseases on CT imaging can be integrated with clinical features as part of a multidisciplinary approach for disease diagnosis.
Collapse
Affiliation(s)
- Linda Le
- Department of Radiology, NYU Langone Health; NYU Grossman School of Medicine, New York, NY
| | - Navneet Narula
- Department of Pathology, NYU Langone Health; NYU Grossman School of Medicine, New York, NY
| | - Fang Zhou
- Department of Pathology, NYU Langone Health; NYU Grossman School of Medicine, New York, NY
| | - Paul Smereka
- Department of Radiology, NYU Langone Health; NYU Grossman School of Medicine, New York, NY
| | - Jeffrey Ordner
- Department of Pathology, NYU Langone Health; NYU Grossman School of Medicine, New York, NY
| | - Neil Theise
- Department of Pathology, NYU Langone Health; NYU Grossman School of Medicine, New York, NY
| | - William H Moore
- Department of Radiology, NYU Langone Health; NYU Grossman School of Medicine, New York, NY
| | - Francis Girvin
- Department of Diagnostic Radiology, Weill Cornell Medicine, New York, NY
| | - Lea Azour
- Department of Radiology, NYU Langone Health; NYU Grossman School of Medicine, New York, NY; Department of Radiological Sciences, UCLA David Geffen School of Medicine, Los Angeles, CA
| | - Andre L Moreira
- Department of Pathology, NYU Langone Health; NYU Grossman School of Medicine, New York, NY
| | - David P Naidich
- Department of Radiology, NYU Langone Health; NYU Grossman School of Medicine, New York, NY
| | - Jane P Ko
- Department of Radiology, NYU Langone Health; NYU Grossman School of Medicine, New York, NY.
| |
Collapse
|
4
|
Rayat Pisheh H, Nojabaei FS, Darvishi A, Rayat Pisheh A, Sani M. Cardiac tissue engineering: an emerging approach to the treatment of heart failure. Front Bioeng Biotechnol 2024; 12:1441933. [PMID: 39211011 PMCID: PMC11357970 DOI: 10.3389/fbioe.2024.1441933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Heart failure is a major health problem in which the heart is unable to pump enough blood to meet the body's needs. It is a progressive disease that becomes more severe over time and can be caused by a variety of factors, including heart attack, cardiomyopathy and heart valve disease. There are various methods to cure this disease, which has many complications and risks. The advancement of knowledge and technology has proposed new methods for many diseases. One of the promising new treatments for heart failure is tissue engineering. Tissue engineering is a field of research that aims to create living tissues and organs to replace damaged or diseased tissue. The goal of tissue engineering in heart failure is to improve cardiac function and reduce the need for heart transplantation. This can be done using the three important principles of cells, biomaterials and signals to improve function or replace heart tissue. The techniques for using cells and biomaterials such as electrospinning, hydrogel synthesis, decellularization, etc. are diverse. Treating heart failure through tissue engineering is still under development and research, but it is hoped that there will be no transplants or invasive surgeries in the near future. In this study, based on the most important research in recent years, we will examine the power of tissue engineering in the treatment of heart failure.
Collapse
Affiliation(s)
- Hossein Rayat Pisheh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Sadat Nojabaei
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Science, Tehran, Iran
| | - Ahmad Darvishi
- School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Rayat Pisheh
- Department of Biology, Payam Noor University (PUN), Shiraz, Iran
| | - Mahsa Sani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
5
|
Yang J, Lei W, Xiao Y, Tan S, Yang J, Lin Y, Yang Z, Zhao D, Zhang C, Shen Z, Hu S. Generation of human vascularized and chambered cardiac organoids for cardiac disease modelling and drug evaluation. Cell Prolif 2024; 57:e13631. [PMID: 38453465 PMCID: PMC11294415 DOI: 10.1111/cpr.13631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/17/2024] [Accepted: 02/26/2024] [Indexed: 03/09/2024] Open
Abstract
Human induced pluripotent stem cell (hiPSC)-derived cardiac organoids (COs) have shown great potential in modelling human heart development and cardiovascular diseases, a leading cause of global death. However, several limitations such as low reproducibility, limited vascularization and difficulty in formation of cardiac chamber were yet to be overcome. We established a new method for robust generation of COs, via combination of methodologies of hiPSC-derived vascular spheres and directly differentiated cardiomyocytes from hiPSCs, and investigated the potential application of human COs in cardiac injury modelling and drug evaluation. The human COs we built displayed a vascularized and chamber-like structure, and hence were named vaschamcardioids (vcCOs). These vcCOs exhibited approximately 90% spontaneous beating ratio. Single-cell transcriptomics identified a total of six cell types in the vcCOs, including cardiomyocytes, cardiac precursor cells, endothelial cells, fibroblasts, etc. We successfully recaptured the processes of cardiac injury and fibrosis in vivo on vcCOs, and showed that the FDA-approved medication captopril significantly attenuated cardiac injury-induced fibrosis and functional disorders. In addition, the human vcCOs exhibited an obvious drug toxicity reaction to doxorubicin in a dose-dependent manner. We developed a three-step method for robust generation of chamber-like and vascularized complex vcCOs, and our data suggested that vcCOs might become a useful model for understanding pathophysiological mechanisms of cardiovascular diseases, developing intervention strategies and screening drugs.
Collapse
Affiliation(s)
- Jingsi Yang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical College, Soochow UniversitySuzhouChina
| | - Wei Lei
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical College, Soochow UniversitySuzhouChina
| | - Yang Xiao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical College, Soochow UniversitySuzhouChina
| | - Shuai Tan
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical College, Soochow UniversitySuzhouChina
| | - Jiani Yang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical College, Soochow UniversitySuzhouChina
| | - Yingjiong Lin
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical College, Soochow UniversitySuzhouChina
| | - Zhuangzhuang Yang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical College, Soochow UniversitySuzhouChina
| | - Dandan Zhao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical College, Soochow UniversitySuzhouChina
| | - Chunxiang Zhang
- Department of Cardiology, Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, the Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical College, Soochow UniversitySuzhouChina
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and ProtectionSuzhou Medical College, Soochow UniversitySuzhouChina
| |
Collapse
|
6
|
Cimini M, Hansmann UHE, Gonzalez C, Chesney AD, Truongcao MM, Gao E, Wang T, Roy R, Forte E, Mallaredy V, Thej C, Magadum A, Joladarashi D, Benedict C, Koch WJ, Tükel Ç, Kishore R. Podoplanin Positive Cell-derived Extracellular Vesicles Contribute to Cardiac Amyloidosis After Myocardial Infarction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601297. [PMID: 39005419 PMCID: PMC11244852 DOI: 10.1101/2024.06.28.601297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Background Amyloidosis is a major long-term complication of chronic disease; however, whether it represents one of the complications of post-myocardial infarction (MI) is yet to be fully understood. Methods Using wild-type and knocked-out MI mouse models and characterizing in vitro the exosomal communication between bone marrow-derived macrophages and activated mesenchymal stromal cells (MSC) isolated after MI, we investigated the mechanism behind Serum Amyloid A 3 (SAA3) protein overproduction in injured hearts. Results Here, we show that amyloidosis occurs after MI and that amyloid fibers are composed of macrophage-derived SAA3 monomers. SAA3 overproduction in macrophages is triggered by exosomal communication from a subset of activated MSC, which, in response to MI, acquire the expression of a platelet aggregation-inducing type I transmembrane glycoprotein named Podoplanin (PDPN). Cardiac MSC PDPN+ communicate with and activate macrophages through their extracellular vesicles or exosomes. Specifically, MSC PDPN+ derived exosomes (MSC PDPN+ Exosomes) are enriched in SAA3 and exosomal SAA3 protein engages with Toll-like receptor 2 (TRL2) on macrophages, triggering an overproduction and impaired clearance of SAA3 proteins, resulting in aggregation of SAA3 monomers as rigid amyloid deposits in the extracellular space. The onset of amyloid fibers deposition alongside extra-cellular-matrix (ECM) proteins in the ischemic heart exacerbates the rigidity and stiffness of the scar, hindering the contractility of viable myocardium and overall impairing organ function. Using SAA3 and TLR2 deficient mouse models, we show that SAA3 delivered by MSC PDPN+ exosomes promotes post-MI amyloidosis. Inhibition of SAA3 aggregation via administration of a retro-inverso D-peptide, specifically designed to bind SAA3 monomers, prevents the deposition of SAA3 amyloid fibrils, positively modulates the scar formation, and improves heart function post-MI. Conclusion Overall, our findings provide mechanistic insights into post-MI amyloidosis and suggest that SAA3 may be an attractive target for effective scar reversal after ischemic injury and a potential target in multiple diseases characterized by a similar pattern of inflammation and amyloid deposition. NOVELTY AND SIGNIFICANCE What is known? Accumulation of rigid amyloid structures in the left ventricular wall impairs ventricle contractility.After myocardial infarction cardiac Mesenchymal Stromal Cells (MSC) acquire Podoplanin (PDPN) to better interact with immune cells.Amyloid structures can accumulate in the heart after chronic inflammatory conditions. What information does this article contribute? Whether accumulation of cumbersome amyloid structures in the ischemic scar impairs left ventricle contractility, and scar reversal after myocardial infarction (MI) has never been investigated.The pathophysiological relevance of PDPN acquirement by MSC and the functional role of their secreted exosomes in the context of post-MI cardiac remodeling has not been investigated.Amyloid structures are present in the scar after ischemia and are composed of macrophage-derived Serum Amyloid A (SAA) 3 monomers, although mechanisms of SAA3 overproduction is not established. SUMMARY OF NOVELTY AND SIGNIFICANCE Here, we report that amyloidosis, a secondary phenomenon of an already preexisting and prolonged chronic inflammatory condition, occurs after MI and that amyloid structures are composed of macrophage-derived SAA3 monomers. Frequently studied cardiac amyloidosis are caused by aggregation of immunoglobulin light chains, transthyretin, fibrinogen, and apolipoprotein in a healthy heart as a consequence of systemic chronic inflammation leading to congestive heart failure with various types of arrhythmias and tissue stiffness. Although chronic MI is considered a systemic inflammatory condition, studies regarding the possible accumulation of amyloidogenic proteins after MI and the mechanisms involved in that process are yet to be reported. Here, we show that SAA3 overproduction in macrophages is triggered in a Toll-like Receptor 2 (TLR2)-p38MAP Kinase-dependent manner by exosomal communication from a subset of activated MSC, which, in response to MI, express a platelet aggregation-inducing type I transmembrane glycoprotein named Podoplanin. We provide the full mechanism of this phenomenon in murine models and confirm SAA3 amyloidosis in failing human heart samples. Moreover, we developed a retro-inverso D-peptide therapeutic approach, "DRI-R5S," specifically designed to bind SAA3 monomers and prevent post-MI aggregation and deposition of SAA3 amyloid fibrils without interfering with the innate immune response.
Collapse
|
7
|
Poto R, Marone G, Galli SJ, Varricchi G. Mast cells: a novel therapeutic avenue for cardiovascular diseases? Cardiovasc Res 2024; 120:681-698. [PMID: 38630620 PMCID: PMC11135650 DOI: 10.1093/cvr/cvae066] [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: 07/26/2023] [Revised: 11/28/2023] [Accepted: 01/08/2024] [Indexed: 04/19/2024] Open
Abstract
Mast cells are tissue-resident immune cells strategically located in different compartments of the normal human heart (the myocardium, pericardium, aortic valve, and close to nerves) as well as in atherosclerotic plaques. Cardiac mast cells produce a broad spectrum of vasoactive and proinflammatory mediators, which have potential roles in inflammation, angiogenesis, lymphangiogenesis, tissue remodelling, and fibrosis. Mast cells release preformed mediators (e.g. histamine, tryptase, and chymase) and de novo synthesized mediators (e.g. cysteinyl leukotriene C4 and prostaglandin D2), as well as cytokines and chemokines, which can activate different resident immune cells (e.g. macrophages) and structural cells (e.g. fibroblasts and endothelial cells) in the human heart and aorta. The transcriptional profiles of various mast cell populations highlight their potential heterogeneity and distinct gene and proteome expression. Mast cell plasticity and heterogeneity enable these cells the potential for performing different, even opposite, functions in response to changing tissue contexts. Human cardiac mast cells display significant differences compared with mast cells isolated from other organs. These characteristics make cardiac mast cells intriguing, given their dichotomous potential roles of inducing or protecting against cardiovascular diseases. Identification of cardiac mast cell subpopulations represents a prerequisite for understanding their potential multifaceted roles in health and disease. Several new drugs specifically targeting human mast cell activation are under development or in clinical trials. Mast cells and/or their subpopulations can potentially represent novel therapeutic targets for cardiovascular disorders.
Collapse
Affiliation(s)
- Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- World Allergy Organization (WAO), Center of Excellence (CoE), Via S. Pansini 5, Naples 80131, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- World Allergy Organization (WAO), Center of Excellence (CoE), Via S. Pansini 5, Naples 80131, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- Institute of Experimental Endocrinology and Oncology ‘G. Salvatore’, National Research Council (CNR), Via S. Pansini 5, Naples 80131, Italy
| | - Stephen J Galli
- Department of Pathology and the Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, 291 Campus Dr, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, 291 Campus Dr, Stanford, CA, USA
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- World Allergy Organization (WAO), Center of Excellence (CoE), Via S. Pansini 5, Naples 80131, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
- Institute of Experimental Endocrinology and Oncology ‘G. Salvatore’, National Research Council (CNR), Via S. Pansini 5, Naples 80131, Italy
| |
Collapse
|
8
|
Vargas Aguilar S, Cui M, Tan W, Sanchez-Ortiz E, Bassel-Duby R, Liu N, Olson EN. The PD-1-PD-L1 pathway maintains an immunosuppressive environment essential for neonatal heart regeneration. NATURE CARDIOVASCULAR RESEARCH 2024; 3:389-402. [PMID: 38737787 PMCID: PMC11086661 DOI: 10.1038/s44161-024-00447-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 02/05/2024] [Indexed: 05/14/2024]
Abstract
The adult mouse heart responds to injury by scarring with consequent loss of contractile function, whereas the neonatal heart possesses the ability to regenerate. Activation of the immune system is among the first events upon tissue injury. It has been shown that immune response kinetics differ between regeneration and pathological remodeling, yet the underlying mechanisms of the distinct immune reactions during tissue healing remain unclear. Here we show that the immunomodulatory PD-1-PD-L1 pathway is highly active in regenerative neonatal hearts but rapidly silenced later in life. Deletion of the PD-1 receptor or inactivation of its ligand PD-L1 prevented regeneration of neonatal hearts after injury. Disruption of the pathway during neonatal cardiac injury led to increased inflammation and aberrant T cell activation, which ultimately impaired cardiac regeneration. Our findings reveal an immunomodulatory and cardioprotective role for the PD-1-PD-L1 pathway in heart regeneration and offer potential avenues for the control of adult tissue regeneration.
Collapse
Affiliation(s)
- Stephanie Vargas Aguilar
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- These authors contributed equally: Stephanie Vargas Aguilar, Miao Cui
| | - Miao Cui
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Cardiology, Boston Children's Hospital and Department of Genetics, Harvard Medical School, Boston, MA, USA
- These authors contributed equally: Stephanie Vargas Aguilar, Miao Cui
| | - Wei Tan
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Efrain Sanchez-Ortiz
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ning Liu
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eric N Olson
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
9
|
Picchio V, Gaetani R, Pagano F, Derevyanchuk Y, Pagliarosi O, Floris E, Cozzolino C, Bernava G, Bordin A, Rocha F, Pereira ARS, Ministro A, Pinto AT, De Falco E, Serino G, Massai D, Tamarat R, Pesce M, Santos SCR, Messina E, Chimenti I. Early Impairment of Paracrine and Phenotypic Features in Resident Cardiac Mesenchymal Stromal Cells after Thoracic Radiotherapy. Int J Mol Sci 2024; 25:2873. [PMID: 38474123 DOI: 10.3390/ijms25052873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Radiotherapy-induced cardiac toxicity and consequent diseases still represent potential severe late complications for many cancer survivors who undergo therapeutic thoracic irradiation. We aimed to assess the phenotypic and paracrine features of resident cardiac mesenchymal stromal cells (CMSCs) at early follow-up after the end of thoracic irradiation of the heart as an early sign and/or mechanism of cardiac toxicity anticipating late organ dysfunction. Resident CMSCs were isolated from a rat model of fractionated thoracic irradiation with accurate and clinically relevant heart dosimetry that developed delayed dose-dependent cardiac dysfunction after 1 year. Cells were isolated 6 and 12 weeks after the end of radiotherapy and fully characterized at the transcriptional, paracrine, and functional levels. CMSCs displayed several altered features in a dose- and time-dependent trend, with the most impaired characteristics observed in those exposed in situ to the highest radiation dose with time. In particular, altered features included impaired cell migration and 3D growth and a and significant association of transcriptomic data with GO terms related to altered cytokine and growth factor signaling. Indeed, the altered paracrine profile of CMSCs derived from the group at the highest dose at the 12-week follow-up gave significantly reduced angiogenic support to endothelial cells and polarized macrophages toward a pro-inflammatory profile. Data collected in a clinically relevant rat model of heart irradiation simulating thoracic radiotherapy suggest that early paracrine and transcriptional alterations of the cardiac stroma may represent a dose- and time-dependent biological substrate for the delayed cardiac dysfunction phenotype observed in vivo.
Collapse
Affiliation(s)
- Vittorio Picchio
- Department of Angio Cardio Neurology, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Roberto Gaetani
- Department of Molecular Medicine, Sapienza University, 00161 Roma, Italy
| | - Francesca Pagano
- Institute of Biochemistry and Cell Biology, National Council of Research (IBBC-CNR), 00015 Monterotondo, Italy
| | - Yuriy Derevyanchuk
- Department of Molecular Medicine, Sapienza University, 00161 Roma, Italy
| | - Olivia Pagliarosi
- Department of Molecular Medicine, Sapienza University, 00161 Roma, Italy
| | - Erica Floris
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy
| | - Claudia Cozzolino
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy
| | | | - Antonella Bordin
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy
| | - Filipe Rocha
- Centro Cardiovascular da Universidade de Lisboa (CCUL@RISE), Lisbon School of Medicine, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Ana Rita Simões Pereira
- Centro Cardiovascular da Universidade de Lisboa (CCUL@RISE), Lisbon School of Medicine, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Augusto Ministro
- Centro Cardiovascular da Universidade de Lisboa (CCUL@RISE), Lisbon School of Medicine, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Ana Teresa Pinto
- Centro Cardiovascular da Universidade de Lisboa (CCUL@RISE), Lisbon School of Medicine, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Elena De Falco
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy
- Mediterranea Cardiocentro, 80122 Napoli, Italy
| | - Gianpaolo Serino
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Torino, Italy
| | - Diana Massai
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Torino, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, 10129 Torino, Italy
| | - Radia Tamarat
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), 92260 Fontenay-aux-Roses, France
| | | | - Susana Constantino Rosa Santos
- Centro Cardiovascular da Universidade de Lisboa (CCUL@RISE), Lisbon School of Medicine, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Elisa Messina
- Department of Molecular Medicine, Sapienza University, 00161 Roma, Italy
| | - Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy
- Mediterranea Cardiocentro, 80122 Napoli, Italy
| |
Collapse
|
10
|
Heusch G. Myocardial ischemia/reperfusion: Translational pathophysiology of ischemic heart disease. MED 2024; 5:10-31. [PMID: 38218174 DOI: 10.1016/j.medj.2023.12.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/27/2023] [Accepted: 12/12/2023] [Indexed: 01/15/2024]
Abstract
Ischemic heart disease is the greatest health burden and most frequent cause of death worldwide. Myocardial ischemia/reperfusion is the pathophysiological substrate of ischemic heart disease. Improvements in prevention and treatment of ischemic heart disease have reduced mortality in developed countries over the last decades, but further progress is now stagnant, and morbidity and mortality from ischemic heart disease in developing countries are increasing. Significant problems remain to be resolved and require a better pathophysiological understanding. The present review attempts to briefly summarize the state of the art in myocardial ischemia/reperfusion research, with a view on both its coronary vascular and myocardial aspects, and to define the cutting edges where further mechanistic knowledge is needed to facilitate translation to clinical practice.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
11
|
Qiao B, Liu X, Wang B, Wei S. The role of periostin in cardiac fibrosis. Heart Fail Rev 2024; 29:191-206. [PMID: 37870704 DOI: 10.1007/s10741-023-10361-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/10/2023] [Indexed: 10/24/2023]
Abstract
Cardiac fibrosis, which is the buildup of proteins in the connective tissues of the heart, can lead to end-stage extracellular matrix (ECM) remodeling and ultimately heart failure. Cardiac remodeling involves changes in gene expression in cardiac cells and ECM, which significantly leads to the morbidity and mortality in heart failure. However, despite extensive research, the elusive intricacies underlying cardiac fibrosis remain unidentified. Periostin, an extracellular matrix (ECM) protein of the fasciclin superfamily, acts as a scaffold for building complex architectures in the ECM, which improves intermolecular interactions and augments the mechanical properties of connective tissues. Recent research has shown that periostin not only contributes to normal ECM homeostasis in a healthy heart but also serves as a potent inducible regulator of cellular reorganization in cardiac fibrosis. Here, we reviewed the constitutive domain of periostin and its interaction with other ECM proteins. We have also discussed the critical pathophysiological functions of periostin in cardiac remodeling mechanisms, including two distinct yet potentially intertwined mechanisms. Furthermore, we will focus on the intrinsic complexities within periostin research, particularly surrounding the contentious issues observed in experimental findings.
Collapse
Affiliation(s)
- Bao Qiao
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Xuehao Liu
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Bailu Wang
- Clinical Trial Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Shujian Wei
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
12
|
Kourampi I, Katsioupa M, Oikonomou E, Tsigkou V, Marinos G, Goliopoulou A, Katsarou O, Kalogeras K, Theofilis P, Tsatsaragkou A, Siasos G, Tousoulis D, Vavuranakis M. The Role of Ranolazine in Heart Failure-Current Concepts. Am J Cardiol 2023; 209:92-103. [PMID: 37844876 DOI: 10.1016/j.amjcard.2023.09.066] [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: 07/08/2023] [Revised: 09/09/2023] [Accepted: 09/15/2023] [Indexed: 10/18/2023]
Abstract
Heart failure is a complex clinical syndrome with a detrimental impact on mortality and morbidity. Energy substrate utilization and myocardial ion channel regulation have gained research interest especially after the introduction of sodium-glucose co-transporter 2 inhibitors in the treatment of heart failure. Ranolazine or N-(2,6-dimethylphenyl)-2-(4-[2-hydroxy-3-(2-methoxyphenoxy) propyl] piperazin-1-yl) acetamide hydrochloride is an active piperazine derivative which inhibits late sodium current thus minimizing calcium overload in the ischemic cardiomyocytes. Ranolazine also prevents fatty acid oxidation and favors glycose utilization ameliorating the "energy starvation" of the failing heart. Heart failure with preserved ejection fraction is characterized by diastolic impairment; according to the literature ranolazine could be beneficial in the management of increased left ventricular end-diastolic pressure, right ventricular systolic dysfunction and wall shear stress which is reflected by the high natriuretic peptides. Fewer data is evident regarding the effects of ranolazine in heart failure with reduced ejection fraction and mainly support the control of the sodium-calcium exchanger and function of sarcoendoplasmic reticulum calcium adenosine triphosphatase. Ranolazine's therapeutic mechanisms in myocardial ion channels and energy utilization are documented in patients with chronic coronary syndromes. Nevertheless, ranolazine might have a broader effect in the therapy of heart failure and further mechanistic research is required.
Collapse
Affiliation(s)
- Islam Kourampi
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Maria Katsioupa
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece.
| | - Vasiliki Tsigkou
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Georgios Marinos
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Athina Goliopoulou
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Ourania Katsarou
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Konstantinos Kalogeras
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Panagiotis Theofilis
- 1st Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Aikaterini Tsatsaragkou
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece; Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston Massachusetts
| | - Dimitris Tousoulis
- 1st Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Manolis Vavuranakis
- 3rd Department of Cardiology, 'Sotiria' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
13
|
Volmert B, Kiselev A, Juhong A, Wang F, Riggs A, Kostina A, O'Hern C, Muniyandi P, Wasserman A, Huang A, Lewis-Israeli Y, Panda V, Bhattacharya S, Lauver A, Park S, Qiu Z, Zhou C, Aguirre A. A patterned human primitive heart organoid model generated by pluripotent stem cell self-organization. Nat Commun 2023; 14:8245. [PMID: 38086920 PMCID: PMC10716495 DOI: 10.1038/s41467-023-43999-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Pluripotent stem cell-derived organoids can recapitulate significant features of organ development in vitro. We hypothesized that creating human heart organoids by mimicking aspects of in utero gestation (e.g., addition of metabolic and hormonal factors) would lead to higher physiological and anatomical relevance. We find that heart organoids produced using this self-organization-driven developmental induction strategy are remarkably similar transcriptionally and morphologically to age-matched human embryonic hearts. We also show that they recapitulate several aspects of cardiac development, including large atrial and ventricular chambers, proepicardial organ formation, and retinoic acid-mediated anterior-posterior patterning, mimicking the developmental processes found in the post-heart tube stage primitive heart. Moreover, we provide proof-of-concept demonstration of the value of this system for disease modeling by exploring the effects of ondansetron, a drug administered to pregnant women and associated with congenital heart defects. These findings constitute a significant technical advance for synthetic heart development and provide a powerful tool for cardiac disease modeling.
Collapse
Affiliation(s)
- Brett Volmert
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Artem Kiselev
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, USA
- Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- Division of Dermatology, Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Aniwat Juhong
- Institute for Quantitative Health Science and Engineering, Division of Biomedical Devices, Michigan State University, East Lansing, MI, USA
- Department of Electrical and Computer Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Fei Wang
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Ashlin Riggs
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Aleksandra Kostina
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Colin O'Hern
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Priyadharshni Muniyandi
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Aaron Wasserman
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Amanda Huang
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Yonatan Lewis-Israeli
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Vishal Panda
- Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Division of Systems Biology, Michigan State University, East Lansing, MI, USA
| | - Sudin Bhattacharya
- Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Division of Systems Biology, Michigan State University, East Lansing, MI, USA
| | - Adam Lauver
- Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Sangbum Park
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, USA
- Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- Division of Dermatology, Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Zhen Qiu
- Institute for Quantitative Health Science and Engineering, Division of Biomedical Devices, Michigan State University, East Lansing, MI, USA
- Department of Electrical and Computer Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA
| | - Chao Zhou
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Aitor Aguirre
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, USA.
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
14
|
Hardy SA, Liesinger L, Patrick R, Poettler M, Rech L, Gindlhuber J, Mabotuwana NS, Ashour D, Stangl V, Bigland M, Murtha LA, Starkey MR, Scherr D, Hansbro PM, Hoefler G, Campos Ramos G, Cochain C, Harvey RP, Birner-Gruenberger R, Boyle AJ, Rainer PP. Extracellular Matrix Protein-1 as a Mediator of Inflammation-Induced Fibrosis After Myocardial Infarction. JACC Basic Transl Sci 2023; 8:1539-1554. [PMID: 38205347 PMCID: PMC10774582 DOI: 10.1016/j.jacbts.2023.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 05/17/2023] [Accepted: 05/17/2023] [Indexed: 01/12/2024]
Abstract
Irreversible fibrosis is a hallmark of myocardial infarction (MI) and heart failure. Extracellular matrix protein-1 (ECM-1) is up-regulated in these hearts, localized to fibrotic, inflammatory, and perivascular areas. ECM-1 originates predominantly from fibroblasts, macrophages, and pericytes/vascular cells in uninjured human and mouse hearts, and from M1 and M2 macrophages and myofibroblasts after MI. ECM-1 stimulates fibroblast-to-myofibroblast transition, up-regulates key fibrotic and inflammatory pathways, and inhibits cardiac fibroblast migration. ECM-1 binds HuCFb cell surface receptor LRP1, and LRP1 inhibition blocks ECM-1 from stimulating fibroblast-to-myofibroblast transition, confirming a novel ECM-1-LRP1 fibrotic signaling axis. ECM-1 may represent a novel mechanism facilitating inflammation-fibrosis crosstalk.
Collapse
Affiliation(s)
- Sean A. Hardy
- Department of Internal Medicine and University Heart Center, Division of Cardiology, Medical University of Graz, Graz, Austria
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Laura Liesinger
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
- Institute of Chemical Technologies and Analytical Chemistry, Technische Universität Wien, Vienna, Austria
| | - Ralph Patrick
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Maria Poettler
- Department of Internal Medicine and University Heart Center, Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Lavinia Rech
- Department of Internal Medicine and University Heart Center, Division of Cardiology, Medical University of Graz, Graz, Austria
- Department of Cardiac Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Nishani S. Mabotuwana
- Department of Internal Medicine and University Heart Center, Division of Cardiology, Medical University of Graz, Graz, Austria
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - DiyaaEldin Ashour
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Verena Stangl
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Mark Bigland
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Lucy A. Murtha
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Malcolm R. Starkey
- Department of Immunology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Daniel Scherr
- Department of Internal Medicine and University Heart Center, Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Philip M. Hansbro
- Centre for Inflammation, Centenary Institute, and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, New South Wales, Australia
| | - Gerald Hoefler
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Gustavo Campos Ramos
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
- Department of Internal Medicine 1, University Hospital of Würzburg, Würzburg, Germany
| | - Clement Cochain
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Richard P. Harvey
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, UNSW Sydney, Sydney, Australia
| | - Ruth Birner-Gruenberger
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
- Institute of Chemical Technologies and Analytical Chemistry, Technische Universität Wien, Vienna, Austria
- BioTechMed Graz, Graz, Austria
| | - Andrew J. Boyle
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Department of Cardiovascular Medicine, John Hunter Hospital, New Lambton Heights, New South Wales, Australia
| | - Peter P. Rainer
- Department of Internal Medicine and University Heart Center, Division of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Department of Medicine, St. Johann in Tirol General Hospital, St. Johann in Tirol, Austria
| |
Collapse
|
15
|
Miquelestorena-Standley E, da Silva AVV, Monnier M, Chadet S, Piollet M, Héraud A, Lemoine R, Bochaton T, Derumeaux G, Roger S, Ivanes F, Angoulvant D. Human peripheral blood mononuclear cells display a temporal evolving inflammatory profile after myocardial infarction and modify myocardial fibroblasts phenotype. Sci Rep 2023; 13:16745. [PMID: 37798364 PMCID: PMC10556078 DOI: 10.1038/s41598-023-44036-3] [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: 06/03/2023] [Accepted: 10/03/2023] [Indexed: 10/07/2023] Open
Abstract
Pathophysiological response after acute myocardial infarction (AMI) is described as a three-stage model involving temporal phenotypic modifications of both immune cells and fibroblasts: a primary inflammatory phase, followed by a reparative phase and a fibrous scar maturation phase. Purinergic receptors, particularly the P2Y11 receptor, have been reported to be involved in the regulation of inflammation after ischemia and could act for the resolution of inflammation after AMI. For the first time, we characterized the immuno-inflammatory and P2Y11 expression profiles of peripheral blood mononuclear cells (PBMC) from AMI patients and analyzed the consequences of presenting these cells to cardiac fibroblasts in vitro. PBMC from 178 patients were collected at various times after reperfused ST-segment elevation AMI, from H0 to M12. Expression level of P2RY11 and genes involved in tolerogenic profile of dendritic cells and T cell polarization were evaluated by RT-PCR. P2Y11 protein expression was assessed by flow cytometry. PBMC and human cardiac fibroblasts (HCF) were cocultured and α-SMA/vimentin ratio was analyzed by flow cytometry. Within the first 48 h after AMI, expression levels of HMOX1, STAT3 and CD4 increased while IDO1 and TBX21/GATA3 ratio decreased. Concomitantly, the expression of P2RY11 increased in both T and B cells. In vitro, PBMC collected at H48 after AMI induced an increase in α-SMA/vimentin ratio in HCF. Our results suggest that human PBMC display an evolving inflammatory profile with reparative characteristics the first two days after AMI and secrete soluble mediators leading to the fibroblastic proteins modification, thus participating to myocardial fibrosis.
Collapse
Affiliation(s)
- Elodie Miquelestorena-Standley
- EA 4245 Transplantation, Immunologie, Inflammation, Faculté de Médecine, Université de Tours, 10 boulevard tonnele, 37032, Tours Cedex 1, France.
- Service d'Anatomie et Cytologie Pathologiques, CHRU de Tours, Tours, France.
| | - Ana Valéria Vinhais da Silva
- EA 4245 Transplantation, Immunologie, Inflammation, Faculté de Médecine, Université de Tours, 10 boulevard tonnele, 37032, Tours Cedex 1, France
| | - Marina Monnier
- EA 4245 Transplantation, Immunologie, Inflammation, Faculté de Médecine, Université de Tours, 10 boulevard tonnele, 37032, Tours Cedex 1, France
| | - Stéphanie Chadet
- EA 4245 Transplantation, Immunologie, Inflammation, Faculté de Médecine, Université de Tours, 10 boulevard tonnele, 37032, Tours Cedex 1, France
| | - Marie Piollet
- EA 4245 Transplantation, Immunologie, Inflammation, Faculté de Médecine, Université de Tours, 10 boulevard tonnele, 37032, Tours Cedex 1, France
| | - Audrey Héraud
- EA 4245 Transplantation, Immunologie, Inflammation, Faculté de Médecine, Université de Tours, 10 boulevard tonnele, 37032, Tours Cedex 1, France
| | - Roxane Lemoine
- EA 4245 Transplantation, Immunologie, Inflammation, Faculté de Médecine, Université de Tours, 10 boulevard tonnele, 37032, Tours Cedex 1, France
| | - Thomas Bochaton
- Service de Cardiologie, Hospices Civils de Lyon, Lyon, France
| | - Geneviève Derumeaux
- Service de Physiologie, Hôpital Henri Mondor, AP-HP, Université Paris-Est Créteil, INSERM U955, Créteil, France
| | - Sébastien Roger
- EA 4245 Transplantation, Immunologie, Inflammation, Faculté de Médecine, Université de Tours, 10 boulevard tonnele, 37032, Tours Cedex 1, France
| | - Fabrice Ivanes
- EA 4245 Transplantation, Immunologie, Inflammation, Faculté de Médecine, Université de Tours, 10 boulevard tonnele, 37032, Tours Cedex 1, France
- Service de Cardiologie, CHRU de Tours, Tours, France
| | - Denis Angoulvant
- EA 4245 Transplantation, Immunologie, Inflammation, Faculté de Médecine, Université de Tours, 10 boulevard tonnele, 37032, Tours Cedex 1, France
- Service de Cardiologie, CHRU de Tours, Tours, France
| |
Collapse
|
16
|
Hulsmans M, Schloss MJ, Lee IH, Bapat A, Iwamoto Y, Vinegoni C, Paccalet A, Yamazoe M, Grune J, Pabel S, Momin N, Seung H, Kumowski N, Pulous FE, Keller D, Bening C, Green U, Lennerz JK, Mitchell RN, Lewis A, Casadei B, Iborra-Egea O, Bayes-Genis A, Sossalla S, Ong CS, Pierson RN, Aster JC, Rohde D, Wojtkiewicz GR, Weissleder R, Swirski FK, Tellides G, Tolis G, Melnitchouk S, Milan DJ, Ellinor PT, Naxerova K, Nahrendorf M. Recruited macrophages elicit atrial fibrillation. Science 2023; 381:231-239. [PMID: 37440641 PMCID: PMC10448807 DOI: 10.1126/science.abq3061] [Citation(s) in RCA: 44] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/02/2023] [Indexed: 07/15/2023]
Abstract
Atrial fibrillation disrupts contraction of the atria, leading to stroke and heart failure. We deciphered how immune and stromal cells contribute to atrial fibrillation. Single-cell transcriptomes from human atria documented inflammatory monocyte and SPP1+ macrophage expansion in atrial fibrillation. Combining hypertension, obesity, and mitral valve regurgitation (HOMER) in mice elicited enlarged, fibrosed, and fibrillation-prone atria. Single-cell transcriptomes from HOMER mouse atria recapitulated cell composition and transcriptome changes observed in patients. Inhibiting monocyte migration reduced arrhythmia in Ccr2-∕- HOMER mice. Cell-cell interaction analysis identified SPP1 as a pleiotropic signal that promotes atrial fibrillation through cross-talk with local immune and stromal cells. Deleting Spp1 reduced atrial fibrillation in HOMER mice. These results identify SPP1+ macrophages as targets for immunotherapy in atrial fibrillation.
Collapse
Affiliation(s)
- Maarten Hulsmans
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Maximilian J. Schloss
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - I-Hsiu Lee
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Aneesh Bapat
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yoshiko Iwamoto
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Claudio Vinegoni
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Alexandre Paccalet
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Masahiro Yamazoe
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jana Grune
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Steffen Pabel
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Noor Momin
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Hana Seung
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Nina Kumowski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Fadi E. Pulous
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Daniel Keller
- Department of Thoracic and Cardiovascular Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Constanze Bening
- Department of Thoracic and Cardiovascular Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Ursula Green
- Department of Pathology, Center for Integrated Diagnostics, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jochen K. Lennerz
- Department of Pathology, Center for Integrated Diagnostics, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Richard N. Mitchell
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew Lewis
- Radcliffe Department of Medicine, NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
- British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Barbara Casadei
- Radcliffe Department of Medicine, NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
- British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Oriol Iborra-Egea
- Institut del Cor Germans Trias i Pujol, CIBERCV, Badalona, Barcelona, Spain
| | - Antoni Bayes-Genis
- Institut del Cor Germans Trias i Pujol, CIBERCV, Badalona, Barcelona, Spain
| | - Samuel Sossalla
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
- Department of Cardiology and Angiology, University of Giessen/DZHK, Partner Site Rhein-Main, Germany
| | - Chin Siang Ong
- Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Richard N. Pierson
- Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jon C. Aster
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - David Rohde
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Gregory R. Wojtkiewicz
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Filip K. Swirski
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - George Tellides
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - George Tolis
- Department of Cardiac Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Serguei Melnitchouk
- Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Patrick T. Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Kamila Naxerova
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Internal Medicine I, University Hospital Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
17
|
Chimenti I, Gaetani R, Pagano F. Editorial: The cardiac stroma in homeostasis and disease. Front Cardiovasc Med 2023; 10:1248750. [PMID: 37492159 PMCID: PMC10364592 DOI: 10.3389/fcvm.2023.1248750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 06/30/2023] [Indexed: 07/27/2023] Open
Affiliation(s)
- Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, Latina, Italy
- Mediterranea Cardiocentro, Napoli, Italy
| | - Roberto Gaetani
- Department of Molecular Medicine, Sapienza University, Rome, Italy
- Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California, San Diego, CA, United States
| | - Francesca Pagano
- Institute of Biochemistry and Cell Biology, National Council of Research (IBBC-CNR), Monterotondo, Italy
| |
Collapse
|
18
|
Chowkwale M, Lindsey ML, Saucerman JJ. Intercellular model predicts mechanisms of inflammation-fibrosis coupling after myocardial infarction. J Physiol 2023; 601:2635-2654. [PMID: 35862254 PMCID: PMC9859968 DOI: 10.1113/jp283346] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/18/2022] [Indexed: 01/25/2023] Open
Abstract
After myocardial infarction (MI), cardiac cells work together to regulate wound healing of the infarct. The pathological response to MI yields cardiac remodelling comprising inflammatory and fibrosis phases, and the interplay of cellular dynamics that underlies these phases has not been elucidated. This study developed a computational model to identify cytokine and cellular dynamics post-MI to predict mechanisms driving post-MI inflammation, resolution of inflammation, and scar formation. Additionally, this study evaluated the interdependence between inflammation and fibrosis. Our model bypassed limitations of in vivo approaches in achieving cellular specificity and performing specific perturbations such as global knockouts of chemical factors. The model predicted that inflammation is a graded response to initial infarct size that is amplified by a positive feedback loop between neutrophils and interleukin 1β (IL-1β). Resolution of inflammation was driven by degradation of IL-1β, matrix metalloproteinase 9, and transforming growth factor β (TGF-β), as well as apoptosis of neutrophils. Inflammation regulated TGFβ secretion directly through immune cell recruitment and indirectly through upregulation of macrophage phagocytosis. Lastly, we found that mature collagen deposition was an ultrasensitive switch in response to inflammation, which was amplified primarily by cardiac fibroblast proliferation. These findings describe the relationship between inflammation and fibrosis and highlight how the two responses work together post-MI. This model revealed that post-MI inflammation and fibrosis are dynamically coupled, which provides rationale for designing novel anti-inflammatory, pro-resolving or anti-fibrotic therapies that may improve the response to MI. KEY POINTS: Inflammation and matrix remodelling are two processes involved in wound healing after a heart attack. Cardiac cells work together to facilitate these processes; this is done by secreting cytokines that then regulate the cells themselves or other cells surrounding them. This study developed a computational model of the dynamics of cardiac cells and cytokines to predict mechanisms through which inflammation and matrix remodelling is regulated. We show the roles of various cytokines and signalling motifs in driving inflammation, resolution of inflammation and fibrosis. The novel concept of inflammation-fibrosis coupling, based on the model prediction that inflammation and fibrosis are dynamically coupled, provides rationale for future studies and for designing therapeutics to improve the response after a heart attack.
Collapse
Affiliation(s)
- Mukti Chowkwale
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Merry L. Lindsey
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN
- Research Service, Nashville VA Medical Center, Nashville, TN
| | - Jeffrey J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA
| |
Collapse
|
19
|
Bracco Gartner TCL, Wang Y, Leiteris L, van Adrichem I, Marsman J, Goumans MJ, Bouten CVC, Sluijter JPG, den Toonder JMJ, Suyker WJL, Hjortnaes J. Cyclic strain has antifibrotic effects on the human cardiac fibroblast transcriptome in a human cardiac fibrosis-on-a-chip platform. J Mech Behav Biomed Mater 2023; 144:105980. [PMID: 37399762 DOI: 10.1016/j.jmbbm.2023.105980] [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: 02/15/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 07/05/2023]
Abstract
In cardiac fibrosis, in response to stress or injury, cardiac fibroblasts deposit excessive amounts of collagens which contribute to the development of heart failure. The biochemical stimuli in this process have been extensively studied, but the influence of cyclic deformation on the fibrogenic behavior of cardiac fibroblasts in the ever-beating heart is not fully understood. In fact, most investigated mechanotransduction pathways in cardiac fibroblasts seem to ultimately have profibrotic effects, which leaves an important question in cardiac fibrosis research unanswered: how do cardiac fibroblasts stay quiescent in the ever-beating human heart? In this study, we developed a human cardiac fibrosis-on-a-chip platform and utilized it to investigate if and how cyclic strain affects fibrogenic signaling. The pneumatically actuated platform can expose engineered tissues to controlled strain magnitudes of 0-25% - which covers the entire physiological and pathological strain range in the human heart - and to biochemical stimuli and enables high-throughput screening of multiple samples. Microtissues of human fetal cardiac fibroblasts (hfCF) embedded in gelatin methacryloyl (GelMA) were 3D-cultured on this platform and exposed to strain conditions which mimic the healthy human heart. The results provide evidence of an antifibrotic effect of the applied strain conditions on cardiac fibroblast behavior, emphasizing the influence of biomechanical stimuli on the fibrogenic process and giving a detailed overview of the mechanosensitive pathways and genes involved, which can be used in the development of novel therapies against cardiac fibrosis.
Collapse
Affiliation(s)
- Tom C L Bracco Gartner
- Department of Cardiothoracic Surgery, UMC Utrecht, Utrecht, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht, the Netherlands; Experimental Cardiology Laboratory, Department of Cardiology, UMC Utrecht, Utrecht, the Netherlands
| | - Ye Wang
- Department of Cardiothoracic Surgery, UMC Utrecht, Utrecht, the Netherlands; Department of Mechanical Engineering, Technical University Eindhoven, Eindhoven, the Netherlands; Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands.
| | - Laurynas Leiteris
- Department of Cardiothoracic Surgery, UMC Utrecht, Utrecht, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht, the Netherlands; Experimental Cardiology Laboratory, Department of Cardiology, UMC Utrecht, Utrecht, the Netherlands
| | - Iris van Adrichem
- Department of Cardiothoracic Surgery, UMC Utrecht, Utrecht, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht, the Netherlands; Experimental Cardiology Laboratory, Department of Cardiology, UMC Utrecht, Utrecht, the Netherlands
| | - Judith Marsman
- Central Diagnostics Laboratory, UMC Utrecht, Utrecht, the Netherlands
| | - Marie José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Carlijn V C Bouten
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Joost P G Sluijter
- Regenerative Medicine Center Utrecht, Utrecht, the Netherlands; Experimental Cardiology Laboratory, Department of Cardiology, UMC Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Jaap M J den Toonder
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Willem J L Suyker
- Department of Cardiothoracic Surgery, UMC Utrecht, Utrecht, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Jesper Hjortnaes
- Department of Cardiothoracic Surgery, UMC Utrecht, Utrecht, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht, the Netherlands; Department of Cardiothoracic Surgery, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
20
|
Brusini R, Tran NLL, Cailleau C, Domergue V, Nicolas V, Dormont F, Calet S, Cajot C, Jouran A, Lepetre-Mouelhi S, Laloy J, Couvreur P, Varna M. Assessment of Squalene-Adenosine Nanoparticles in Two Rodent Models of Cardiac Ischemia-Reperfusion. Pharmaceutics 2023; 15:1790. [PMID: 37513977 PMCID: PMC10384353 DOI: 10.3390/pharmaceutics15071790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
Reperfusion injuries after a period of cardiac ischemia are known to lead to pathological modifications or even death. Among the different therapeutic options proposed, adenosine, a small molecule with platelet anti-aggregate and anti-inflammatory properties, has shown encouraging results in clinical trials. However, its clinical use is severely limited because of its very short half-life in the bloodstream. To overcome this limitation, we have proposed a strategy to encapsulate adenosine in squalene-based nanoparticles (NPs), a biocompatible and biodegradable lipid. Thus, the aim of this study was to assess, whether squalene-based nanoparticles loaded with adenosine (SQAd NPs) were cardioprotective in a preclinical cardiac ischemia/reperfusion model. Obtained SQAd NPs were characterized in depth and further evaluated in vitro. The NPs were formulated with a size of about 90 nm and remained stable up to 14 days at both 4 °C and room temperature. Moreover, these NPs did not show any signs of toxicity, neither on HL-1, H9c2 cardiac cell lines, nor on human PBMC and, further retained their inhibitory platelet aggregation properties. In a mouse model with experimental cardiac ischemia-reperfusion, treatment with SQAd NPs showed a reduction of the area at risk, as well as of the infarct area, although not statistically significant. However, we noted a significant reduction of apoptotic cells on cardiac tissue from animals treated with the NPs. Further studies would be interesting to understand how and through which mechanisms these nanoparticles act on cardiac cells.
Collapse
Affiliation(s)
- Romain Brusini
- Université Paris-Saclay, Institut Galien Paris-Saclay, CNRS UMR 8612, Pole Biologie-Pharmacie-Chimie, Bâtiment Henri Moissan, 6 Rue d'Arsonval, 91400 Orsay, France
| | - Natalie Lan Linh Tran
- Université Paris-Saclay, Institut Galien Paris-Saclay, CNRS UMR 8612, Pole Biologie-Pharmacie-Chimie, Bâtiment Henri Moissan, 6 Rue d'Arsonval, 91400 Orsay, France
- Namur Nanosafety Centre, Department of Pharmacy, Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 5000 Namur, Belgium
| | - Catherine Cailleau
- Université Paris-Saclay, Institut Galien Paris-Saclay, CNRS UMR 8612, Pole Biologie-Pharmacie-Chimie, Bâtiment Henri Moissan, 6 Rue d'Arsonval, 91400 Orsay, France
| | - Valérie Domergue
- Université Paris-Saclay, Inserm, CNRS, Ingénierie et Plateformes au Service de l'Innovation Thérapeutique, ANIMEX, 17 Avenue des Sciences, 91400 Orsay, France
| | - Valérie Nicolas
- Université Paris-Saclay, Inserm, CNRS, Ingénierie et Plateformes au Service de l'Innovation Thérapeutique, MIPSIT, 17 Avenue des Sciences, 91400 Orsay, France
| | - Flavio Dormont
- Université Paris-Saclay, Institut Galien Paris-Saclay, CNRS UMR 8612, Pole Biologie-Pharmacie-Chimie, Bâtiment Henri Moissan, 6 Rue d'Arsonval, 91400 Orsay, France
| | - Serge Calet
- Holochem, Rue du Moulin de la Canne, 45300 Pithiviers, France
| | - Caroline Cajot
- Quality Assistance S.A, Technoparc de Thudinie 2, 6536 Thuin, Belgium
| | - Albin Jouran
- Quality Assistance S.A, Technoparc de Thudinie 2, 6536 Thuin, Belgium
| | - Sinda Lepetre-Mouelhi
- Université Paris-Saclay, Institut Galien Paris-Saclay, CNRS UMR 8612, Pole Biologie-Pharmacie-Chimie, Bâtiment Henri Moissan, 6 Rue d'Arsonval, 91400 Orsay, France
| | - Julie Laloy
- Namur Nanosafety Centre, Department of Pharmacy, Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 5000 Namur, Belgium
| | - Patrick Couvreur
- Université Paris-Saclay, Institut Galien Paris-Saclay, CNRS UMR 8612, Pole Biologie-Pharmacie-Chimie, Bâtiment Henri Moissan, 6 Rue d'Arsonval, 91400 Orsay, France
| | - Mariana Varna
- Université Paris-Saclay, Institut Galien Paris-Saclay, CNRS UMR 8612, Pole Biologie-Pharmacie-Chimie, Bâtiment Henri Moissan, 6 Rue d'Arsonval, 91400 Orsay, France
| |
Collapse
|
21
|
Xu X, Hua X, Mo H, Hu S, Song J. Single-cell RNA sequencing to identify cellular heterogeneity and targets in cardiovascular diseases: from bench to bedside. Basic Res Cardiol 2023; 118:7. [PMID: 36750503 DOI: 10.1007/s00395-022-00972-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/09/2022] [Accepted: 12/09/2022] [Indexed: 02/09/2023]
Abstract
The mechanisms of cardiovascular diseases (CVDs) remain incompletely elucidated. Single-cell RNA sequencing (scRNA-seq) has enabled the profiling of single-cell transcriptomes at unprecedented resolution and throughput, which is critical for deciphering cardiovascular cellular heterogeneity and underlying disease mechanisms, thereby facilitating the development of therapeutic strategies. In this review, we summarize cellular heterogeneity in cardiovascular homeostasis and diseases as well as the discovery of potential disease targets based on scRNA-seq, and yield new insights into the promise of scRNA-seq technology in precision medicine and clinical application.
Collapse
Affiliation(s)
- Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Xiumeng Hua
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Han Mo
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, 518057, China
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
22
|
Zhu Y, Li K, Zhang Q, Nie Y, Yan T, Shi X, Han D. High-Strength Injectable Hydrogel into Perivascular Interstitial Space Enhances Arterial Adventitial Stress. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:1529-1537. [PMID: 36683534 DOI: 10.1021/acs.langmuir.2c02935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Injectable hydrogels with strong mechanical properties have significant potential for biomedical applications, including the development of electronic skin, intelligent medical robots, as well as tissue engineering. In this study, we report on an injectable hydrogel with notable tensile strength and adhesion properties, achieved through cross-linking thiol-terminated four-arm poly (ethylene glycol) using silver-doped nano-hydroxyapatite, modified with dopamine. Subsequently, the hydrogel was injected in vivo through the perivascular interstitial space of rats. The hydrogel wrapped around the damaged abdominal aortic adventitia, which greatly increases the stress strength of the arterial adventitia. We found that the hydrogel was characterized by excellent biocompatibility, and it induced little immune response over a span of 21 days post-implantation. This simple and minimally invasive vascular protection strategy appears promising for the treatment of vascular diseases, such as abdominal aortic aneurysm (AAA).
Collapse
Affiliation(s)
- Yuting Zhu
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing100190, China
| | - Kai Li
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing100190, China
- College of Life Science, Beijing University of Chinese Medicine, Beijing100029, China
| | - Qiang Zhang
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing100190, China
- Hebei Key Laboratory of Nano-Biotechnology, Yanshan University, Qinhuangdao066004, China
| | - Yifeng Nie
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing100190, China
| | - Tun Yan
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing100190, China
- College of Life Science, Beijing University of Chinese Medicine, Beijing100029, China
| | - Xiaoli Shi
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing100190, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing100049, China
| | - Dong Han
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing100190, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing100049, China
| |
Collapse
|
23
|
Harris NR, Bálint L, Dy DM, Nielsen NR, Méndez HG, Aghajanian A, Caron KM. The ebb and flow of cardiac lymphatics: a tidal wave of new discoveries. Physiol Rev 2023; 103:391-432. [PMID: 35953269 PMCID: PMC9576179 DOI: 10.1152/physrev.00052.2021] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 06/16/2022] [Accepted: 07/18/2022] [Indexed: 12/16/2022] Open
Abstract
The heart is imbued with a vast lymphatic network that is responsible for fluid homeostasis and immune cell trafficking. Disturbances in the forces that regulate microvascular fluid movement can result in myocardial edema, which has profibrotic and proinflammatory consequences and contributes to cardiovascular dysfunction. This review explores the complex relationship between cardiac lymphatics, myocardial edema, and cardiac disease. It covers the revised paradigm of microvascular forces and fluid movement around the capillary as well as the arsenal of preclinical tools and animal models used to model myocardial edema and cardiac disease. Clinical studies of myocardial edema and their prognostic significance are examined in parallel to the recent elegant animal studies discerning the pathophysiological role and therapeutic potential of cardiac lymphatics in different cardiovascular disease models. This review highlights the outstanding questions of interest to both basic scientists and clinicians regarding the roles of cardiac lymphatics in health and disease.
Collapse
Affiliation(s)
- Natalie R Harris
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - László Bálint
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Danielle M Dy
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Natalie R Nielsen
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Hernán G Méndez
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Amir Aghajanian
- Division of Cardiology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
24
|
Defining the timeline of periostin upregulation in cardiac fibrosis following acute myocardial infarction in mice. Sci Rep 2022; 12:21863. [PMID: 36529756 PMCID: PMC9760637 DOI: 10.1038/s41598-022-26035-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
After myocardial infarction (MI), the heart's reparative response to the ischemic insult and the related loss of cardiomyocytes involves cardiac fibrosis, in which the damaged tissue is replaced with a fibrous scar. Although the scar is essential to prevent ventricular wall rupture in the infarction zone, it expands over time to remote, non-infarct areas, significantly increasing the extent of fibrosis and markedly altering cardiac structure. Cardiac function in this scenario deteriorates, thereby increasing the probability of heart failure and the risk of death. Recent works have suggested that the matricellular protein periostin, known to be involved in fibrosis, is a candidate therapeutic target for the regulation of MI-induced fibrosis and remodeling. Different strategies for the genetic manipulation of periostin have been proposed previously, yet those works did not properly address the time dependency between periostin activity and cardiac fibrosis. Our study aimed to fill that gap in knowledge and fully elucidate the explicit timing of cellular periostin upregulation in the infarcted heart to enable the safer and more effective post-MI targeting of periostin-producing cells. Surgical MI was performed in C57BL/6J and BALB/c mice by ligation of the left anterior descending coronary artery. Flow cytometry analyses of cells derived from the infarcted hearts and quantitative real-time PCR of the total cellular RNA revealed that periostin expression increased during days 2-7 and peaked on day 7 post-infarct, regardless of mouse strain. The established timeline for cellular periostin expression in the post-MI heart is a significant milestone toward the development of optimal periostin-targeted gene therapy.
Collapse
|
25
|
Zhou X, Zhang C, Wu X, Hu X, Zhang Y, Wang X, Zheng L, Gao P, Du J, Zheng W, Shang H, Hu K, Jiang Z, Nie Y, Hu S, Xiao RP, Zhu X, Xiong JW. Dusp6 deficiency attenuates neutrophil-mediated cardiac damage in the acute inflammatory phase of myocardial infarction. Nat Commun 2022; 13:6672. [PMID: 36335128 PMCID: PMC9637103 DOI: 10.1038/s41467-022-33631-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 09/27/2022] [Indexed: 11/07/2022] Open
Abstract
Dual-specificity phosphatase 6 (DUSP6) serves a specific and conserved function on the dephosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2). We previously identified Dusp6 as a regenerative repressor during zebrafish heart regeneration, therefore we propose to investigate the role of this repressor in mammalian cardiac repair. Utilizing a rat strain harboring Dusp6 nonsense mutation, rat neutrophil-cardiomyocyte co-culture, bone marrow transplanted rats and neutrophil-specific Dusp6 knockout mice, we find that Dusp6 deficiency improves cardiac outcomes by predominantly attenuating neutrophil-mediated myocardial damage in acute inflammatory phase after myocardial infarction. Mechanistically, Dusp6 is transcriptionally activated by p38-C/EBPβ signaling and acts as an effector for maintaining p-p38 activity by down-regulating pERK and p38-targeting phosphatases DUSP1/DUSP16. Our findings provide robust animal models and novel insights for neutrophil-mediated cardiac damage and demonstrate the potential of DUSP6 as a therapeutic target for post-MI cardiac remodeling and other relevant inflammatory diseases.
Collapse
Affiliation(s)
- Xiaohai Zhou
- grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871 China
| | - Chenyang Zhang
- grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871 China ,grid.11135.370000 0001 2256 9319PKU-Nanjing Institute of Translational Medicine, Nanjing, 211800 China
| | - Xueying Wu
- grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871 China
| | - Xinli Hu
- grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871 China
| | - Yan Zhang
- grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871 China
| | - Xuelian Wang
- grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871 China
| | - Lixia Zheng
- grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871 China
| | - Peng Gao
- grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871 China
| | - Jianyong Du
- grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871 China
| | - Wen Zheng
- grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871 China
| | - Haibao Shang
- grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871 China
| | - Keping Hu
- grid.506261.60000 0001 0706 7839Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193 China
| | - Zhengfan Jiang
- grid.11135.370000 0001 2256 9319School of Life Sciences, Peking University, Beijing, 100871 China
| | - Yu Nie
- grid.506261.60000 0001 0706 7839State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037 China
| | - Shengshou Hu
- grid.506261.60000 0001 0706 7839State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037 China
| | - Rui-Ping Xiao
- grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871 China ,grid.11135.370000 0001 2256 9319PKU-Nanjing Institute of Translational Medicine, Nanjing, 211800 China
| | - Xiaojun Zhu
- grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871 China ,grid.11135.370000 0001 2256 9319PKU-Nanjing Institute of Translational Medicine, Nanjing, 211800 China
| | - Jing-Wei Xiong
- grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100871 China ,grid.11135.370000 0001 2256 9319PKU-Nanjing Institute of Translational Medicine, Nanjing, 211800 China
| |
Collapse
|
26
|
Biocompatibility and Connectivity of Semiconductor Nanostructures for Cardiac Tissue Engineering Applications. Bioengineering (Basel) 2022; 9:bioengineering9110621. [DOI: 10.3390/bioengineering9110621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
Nano- or microdevices, enabling simultaneous, long-term, multisite, cellular recording and stimulation from many excitable cells, are expected to make a strategic turn in basic and applied cardiology (particularly tissue engineering) and neuroscience. We propose an innovative approach aiming to elicit bioelectrical information from the cell membrane using an integrated circuit (IC) bearing a coating of nanowires on the chip surface. Nanowires grow directly on the backend of the ICs, thus allowing on-site amplification of bioelectric signals with uniform and controlled morphology and growth of the NWs on templates. To implement this technology, we evaluated the biocompatibility of silicon and zinc oxide nanowires (NWs), used as a seeding substrate for cells in culture, on two different primary cell lines. Human cardiac stromal cells were used to evaluate the effects of ZnO NWs of different lengths on cell behavior, morphology and growth, while BV-2 microglial-like cells and GH4-C1 neuroendocrine-like cell lines were used to evaluate cell membrane–NW interaction and contact when cultured on Si NWs. As the optimization of the contact between integrated microelectronics circuits and cellular membranes represents a long-standing issue, our technological approach may lay the basis for a new era of devices exploiting the microelectronics’ sensitivity and “smartness” to both improve investigation of biological systems and to develop suitable NW-based systems available for tissue engineering and regenerative medicine.
Collapse
|
27
|
Strohm L, Ubbens H, Münzel T, Daiber A, Daub S. Role of CD40(L)-TRAF signaling in inflammation and resolution-a double-edged sword. Front Pharmacol 2022; 13:995061. [PMID: 36267276 PMCID: PMC9577411 DOI: 10.3389/fphar.2022.995061] [Citation(s) in RCA: 6] [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: 07/15/2022] [Accepted: 09/12/2022] [Indexed: 11/26/2022] Open
Abstract
Cardiovascular diseases (CVD) and cardiovascular risk factors are the leading cause of death in the world today. According to the Global Burden of Disease Study, hypertension together with ischemic heart and cerebrovascular diseases is responsible for approximately 40% of all deaths worldwide. The major pathomechanism underlying almost all CVD is atherosclerosis, an inflammatory disorder of the vascular system. Recent large-scale clinical trials demonstrated that inflammation itself is an independent cardiovascular risk factor. Specific anti-inflammatory therapy could decrease cardiovascular mortality in patients with atherosclerosis (increased markers of inflammation). Inflammation, however, can also be beneficial by conferring so-called resolution, a process that contributes to clearing damaged tissue from cell debris upon cell death and thereby represents an essential step for recovery from, e.g., ischemia/reperfusion damage. Based on these considerations, the present review highlights features of the detrimental inflammatory reactions as well as of the beneficial process of immune cell-triggered resolution. In this context, we discuss the polarization of macrophages to either M1 or M2 phenotype and critically assess the role of the CD40L-CD40-TRAF signaling cascade in atherosclerosis and its potential link to resolution. As CD40L can bind to different cellular receptors, it can initiate a broad range of inflammatory processes that may be detrimental or beneficial. Likewise, the signaling of CD40L downstream of CD40 is mainly determined by activation of TRAF1-6 pathways that again can be detrimental or beneficial. Accordingly, CD40(L)-based therapies may be Janus-faced and require sophisticated fine-tuning in order to promote cardioprotection.
Collapse
Affiliation(s)
- Lea Strohm
- Department of Cardiology, Cardiology I—Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Henning Ubbens
- Department of Cardiology, Cardiology I—Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, Cardiology I—Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, Cardiology I—Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Steffen Daub
- Department of Cardiology, Cardiology I—Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
28
|
Picchio V, Floris E, Derevyanchuk Y, Cozzolino C, Messina E, Pagano F, Chimenti I, Gaetani R. Multicellular 3D Models for the Study of Cardiac Fibrosis. Int J Mol Sci 2022; 23:ijms231911642. [PMID: 36232943 PMCID: PMC9569892 DOI: 10.3390/ijms231911642] [Citation(s) in RCA: 1] [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: 07/27/2022] [Revised: 09/19/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022] Open
Abstract
Ex vivo modelling systems for cardiovascular research are becoming increasingly important in reducing lab animal use and boosting personalized medicine approaches. Integrating multiple cell types in complex setups adds a higher level of significance to the models, simulating the intricate intercellular communication of the microenvironment in vivo. Cardiac fibrosis represents a key pathogenetic step in multiple cardiovascular diseases, such as ischemic and diabetic cardiomyopathies. Indeed, allowing inter-cellular interactions between cardiac stromal cells, endothelial cells, cardiomyocytes, and/or immune cells in dedicated systems could make ex vivo models of cardiac fibrosis even more relevant. Moreover, culture systems with 3D architectures further enrich the physiological significance of such in vitro models. In this review, we provide a summary of the multicellular 3D models for the study of cardiac fibrosis described in the literature, such as spontaneous microtissues, bioprinted constructs, engineered tissues, and organs-on-chip, discussing their advantages and limitations. Important discoveries on the physiopathology of cardiac fibrosis, as well as the screening of novel potential therapeutic molecules, have been reported thanks to these systems. Future developments will certainly increase their translational impact for understanding and modulating mechanisms of cardiac fibrosis even further.
Collapse
Affiliation(s)
- Vittorio Picchio
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy
| | - Erica Floris
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy
| | - Yuriy Derevyanchuk
- Department of Molecular Medicine, Sapienza University, 00161 Rome, Italy
| | - Claudia Cozzolino
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy
| | - Elisa Messina
- Department of Molecular Medicine, Sapienza University, 00161 Rome, Italy
| | - Francesca Pagano
- Institute of Biochemistry and Cell Biology, National Council of Research (IBBC-CNR), 00015 Monterotondo, Italy
| | - Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy
- Mediterranea Cardiocentro, 80122 Napoli, Italy
- Correspondence: ; Tel.: +39-077-3175-7234
| | - Roberto Gaetani
- Department of Molecular Medicine, Sapienza University, 00161 Rome, Italy
| |
Collapse
|
29
|
Cifarelli V, Kuda O, Yang K, Liu X, Gross RW, Pietka TA, Heo GS, Sultan D, Luehmann H, Lesser J, Ross M, Goldberg IJ, Gropler RJ, Liu Y, Abumrad NA. Cardiac immune cell infiltration associates with abnormal lipid metabolism. Front Cardiovasc Med 2022; 9:948332. [PMID: 36061565 PMCID: PMC9428462 DOI: 10.3389/fcvm.2022.948332] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/22/2022] [Indexed: 01/05/2023] Open
Abstract
CD36 mediates the uptake of long-chain fatty acids (FAs), a major energy substrate for the myocardium. Under excessive FA supply, CD36 can cause cardiac lipid accumulation and inflammation while its deletion reduces heart FA uptake and lipid content and increases glucose utilization. As a result, CD36 was proposed as a therapeutic target for obesity-associated heart disease. However, more recent reports have shown that CD36 deficiency suppresses myocardial flexibility in fuel preference between glucose and FAs, impairing tissue energy balance, while CD36 absence in tissue macrophages reduces efferocytosis and myocardial repair after injury. In line with the latter homeostatic functions, we had previously reported that CD36-/- mice have chronic subclinical inflammation. Lipids are important for the maintenance of tissue homeostasis and there is limited information on heart lipid metabolism in CD36 deficiency. Here, we document in the hearts of unchallenged CD36-/- mice abnormalities in the metabolism of triglycerides, plasmalogens, cardiolipins, acylcarnitines, and arachidonic acid, and the altered remodeling of these lipids in response to an overnight fast. The hearts were examined for evidence of inflammation by monitoring the presence of neutrophils and pro-inflammatory monocytes/macrophages using the respective positron emission tomography (PET) tracers, 64Cu-AMD3100 and 68Ga-DOTA-ECL1i. We detected significant immune cell infiltration in unchallenged CD36-/- hearts as compared with controls and immune infiltration was also observed in hearts of mice with cardiomyocyte-specific CD36 deficiency. Together, the data show that the CD36-/- heart is in a non-homeostatic state that could compromise its stress response. Non-invasive immune cell monitoring in humans with partial or total CD36 deficiency could help evaluate the risk of impaired heart remodeling and disease.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States,Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States,*Correspondence: Vincenza Cifarelli,
| | - Ondrej Kuda
- Institute of Physiology, Czech Academy of Sciences, Prague, Czechia
| | - Kui Yang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States,Division of Complex Drug Analysis, Office of Testing and Research, U.S. Food and Drug Administration, St. Louis, MO, United States
| | - Xinping Liu
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Richard W. Gross
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Terri A. Pietka
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Gyu Seong Heo
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Deborah Sultan
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Hannah Luehmann
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Josie Lesser
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Morgan Ross
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Ira J. Goldberg
- Division of Endocrinology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Robert J. Gropler
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States,Yongjian Liu,
| | - Nada A. Abumrad
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States,Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, United States,Nada A. Abumrad,
| |
Collapse
|
30
|
Garoffolo G, Casaburo M, Amadeo F, Salvi M, Bernava G, Piacentini L, Chimenti I, Zaccagnini G, Milcovich G, Zuccolo E, Agrifoglio M, Ragazzini S, Baasansuren O, Cozzolino C, Chiesa M, Ferrari S, Carbonaro D, Santoro R, Manzoni M, Casalis L, Raucci A, Molinari F, Menicanti L, Pagano F, Ohashi T, Martelli F, Massai D, Colombo GI, Messina E, Morbiducci U, Pesce M. Reduction of Cardiac Fibrosis by Interference With YAP-Dependent Transactivation. Circ Res 2022; 131:239-257. [PMID: 35770662 DOI: 10.1161/circresaha.121.319373] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Conversion of cardiac stromal cells into myofibroblasts is typically associated with hypoxia conditions, metabolic insults, and/or inflammation, all of which are predisposing factors to cardiac fibrosis and heart failure. We hypothesized that this conversion could be also mediated by response of these cells to mechanical cues through activation of the Hippo transcriptional pathway. The objective of the present study was to assess the role of cellular/nuclear straining forces acting in myofibroblast differentiation of cardiac stromal cells under the control of YAP (yes-associated protein) transcription factor and to validate this finding using a pharmacological agent that interferes with the interactions of the YAP/TAZ (transcriptional coactivator with PDZ-binding motif) complex with their cognate transcription factors TEADs (TEA domain transcription factors), under high-strain and profibrotic stimulation. METHODS We employed high content imaging, 2-dimensional/3-dimensional culture, atomic force microscopy mapping, and molecular methods to prove the role of cell/nuclear straining in YAP-dependent fibrotic programming in a mouse model of ischemia-dependent cardiac fibrosis and in human-derived primitive cardiac stromal cells. We also tested treatment of cells with Verteporfin, a drug known to prevent the association of the YAP/TAZ complex with their cognate transcription factors TEADs. RESULTS Our experiments suggested that pharmacologically targeting the YAP-dependent pathway overrides the profibrotic activation of cardiac stromal cells by mechanical cues in vitro, and that this occurs even in the presence of profibrotic signaling mediated by TGF-β1 (transforming growth factor beta-1). In vivo administration of Verteporfin in mice with permanent cardiac ischemia reduced significantly fibrosis and morphometric remodeling but did not improve cardiac performance. CONCLUSIONS Our study indicates that preventing molecular translation of mechanical cues in cardiac stromal cells reduces the impact of cardiac maladaptive remodeling with a positive effect on fibrosis.
Collapse
Affiliation(s)
- Gloria Garoffolo
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Manuel Casaburo
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Francesco Amadeo
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Massimo Salvi
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy (M.S., D.C., F. Molinari, D.M., U.M.)
| | - Giacomo Bernava
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Luca Piacentini
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Isotta Chimenti
- Department of Medical Surgical Science and Biotechnology, Sapienza University of Rome (I.C., C.C.).,Mediterranea Cardiocentro, Napoli (I.C.)
| | | | | | - Estella Zuccolo
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Marco Agrifoglio
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università di Milano, Milan, Italy (M.A.)
| | - Sara Ragazzini
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Otgon Baasansuren
- Faculty of Engineering, Hokkaido University, Sapporo, Japan (O.B., T.O.)
| | - Claudia Cozzolino
- Department of Medical Surgical Science and Biotechnology, Sapienza University of Rome (I.C., C.C.)
| | - Mattia Chiesa
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Silvia Ferrari
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Dario Carbonaro
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy (M.S., D.C., F. Molinari, D.M., U.M.)
| | - Rosaria Santoro
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Martina Manzoni
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | | | - Angela Raucci
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Filippo Molinari
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy (M.S., D.C., F. Molinari, D.M., U.M.)
| | | | - Francesca Pagano
- Institute of Biochemistry and Cell Biology, National Council of Research (IBBC-CNR), Monterotondo, Italy (F.P.)
| | - Toshiro Ohashi
- Faculty of Engineering, Hokkaido University, Sapporo, Japan (O.B., T.O.)
| | | | - Diana Massai
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy (M.S., D.C., F. Molinari, D.M., U.M.)
| | - Gualtiero I Colombo
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Elisa Messina
- Department of Pediatrics and Infant Neuropsychiatry. Policlinico Umberto I, Sapienza University of Rome (E.M.)
| | - Umberto Morbiducci
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy (M.S., D.C., F. Molinari, D.M., U.M.)
| | - Maurizio Pesce
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| |
Collapse
|
31
|
Minotti G, Menna P, Camilli M, Salvatorelli E, Levi R. Beyond hypertension: Diastolic dysfunction associated with cancer treatment in the era of cardio-oncology. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 94:365-409. [PMID: 35659376 DOI: 10.1016/bs.apha.2022.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cancer patients are at an increased risk of cardiovascular events. Both old-generation cytostatics/cytotoxics and new-generation "targeted" drugs can in fact damage cardiomyocytes, endothelial cells of veins and arteries, specialized cells of the conduction system, pericardium, and valves. A new discipline, cardio-oncology, has therefore developed with the aim of protecting cancer patients from cardiovascular events, while also providing them with the best possible oncologic treatment. Anthracyclines have long been known to elicit cardiotoxicity that, depending on treatment- or patient-related factors, may progress with a variable velocity toward cardiomyopathy and systolic heart failure. However, early compromise of diastolic function may precede systolic dysfunction, and a progression of early diastolic dysfunction to diastolic rather than systolic heart failure has been documented in long-term cancer survivors. This chapter first describes general notions about hypertension in the cancer patient and then moves on reviewing the pathophysiology and clinical trajectories of diastolic dysfunction, and the molecular mechanisms of anthracycline-induced diastolic dysfunction. Diastolic dysfunction can in fact be caused and/or aggravated by hypertension. Pharmacologic foundations and therapeutic opportunities to prevent or treat diastolic dysfunction before it progresses toward heart failure are also reviewed, with a special emphasis on the mechanisms of action of drugs that raised hopes to treat diastolic dysfunction in the general population (sacubitril/valsartan, guanylyl cyclase activators, phosphodiesterase inhibitors, ranolazine, inhibitors of type-2 sodium-glucose-inked transporter). Cardio-oncologists will be confronted with the risk:benefit ratio of using these drugs in the cancer patient.
Collapse
Affiliation(s)
- Giorgio Minotti
- Department of Medicine, Campus Bio-Medico University and Fondazione Policlinico, Rome, Italy.
| | - Pierantonio Menna
- Department of Health Sciences, Campus Bio-Medico University and Fondazione Policlinico, Rome, Italy
| | - Massimiliano Camilli
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome
| | - Emanuela Salvatorelli
- Department of Medicine, Campus Bio-Medico University and Fondazione Policlinico, Rome, Italy
| | - Roberto Levi
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
32
|
Chimenti I, Picchio V, Pagano F, Schirone L, Schiavon S, D'Ambrosio L, Valenti V, Forte M, di Nonno F, Rubattu S, Peruzzi M, Versaci F, Greco E, Calogero A, De Falco E, Frati G, Sciarretta S. The impact of autophagy modulation on phenotype and survival of cardiac stromal cells under metabolic stress. Cell Death Discov 2022; 8:149. [PMID: 35365624 PMCID: PMC8975847 DOI: 10.1038/s41420-022-00924-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 01/31/2022] [Accepted: 02/17/2022] [Indexed: 01/18/2023] Open
Abstract
Cardiac stromal cells (CSCs) embrace multiple phenotypes and are a contributory factor in tissue homeostasis and repair. They can be exploited as therapeutic mediators against cardiac fibrosis and remodeling, but their survival and cardioprotective properties can be decreased by microenvironmental cues. We evaluated the impact of autophagy modulation by different pharmacological/genetic approaches on the viability and phenotype of murine CSCs, which had been subjected to nutrient deprivation or hyperglycemia, in order to mimic relevant stress conditions and risk factors of cardiovascular diseases. Our results show that autophagy is activated in CSCs by nutrient deprivation, and that autophagy induction by trehalose or autophagy-related protein 7 (ATG7)-overexpression can significantly preserve CSC viability. Furthermore, autophagy induction is associated with a higher proportion of primitive, non-activated stem cell antigen 1 (Sca1)-positive cells, and with a reduced fibrotic fraction (positive for the discoidin domain-containing receptor 2, DDR2) in the CSC pool after nutrient deprivation. Hyperglycemia, on the other hand, is associated with reduced autophagic flux in CSCs, and with a significant reduction in primitive Sca1+ cells. Autophagy induction by adenoviral-mediated ATG7-overexpression maintains a cardioprotective, anti-inflammatory and pro-angiogenic paracrine profile of CSCs exposed to hyperglycemia for 1 week. Finally, autophagy induction by ATG7-overexpression during hyperglycemia can significantly preserve cell viability in CSCs, which were subsequently exposed to nutrient deprivation, reducing hyperglycemia-induced impairment of cell resistance to stress. In conclusion, our results show that autophagy stimulation preserves CSC viability and function in response to metabolic stressors, suggesting that it may boost the beneficial functions of CSCs in cardiac repair mechanisms.
Collapse
Affiliation(s)
- Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.
- Mediterranea Cardiocentro, Napoli, Italy.
| | - Vittorio Picchio
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Francesca Pagano
- Biochemistry and Cellular Biology Istitute, CNR, Monterotondo, Italy
| | - Leonardo Schirone
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Department of Clinical, Internal Medicine, Anaesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Sonia Schiavon
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Luca D'Ambrosio
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Valentina Valenti
- Haemodynamic and Cardiology Unit, "Santa Maria Goretti" Hospital, Latina, Italy
| | | | | | - Speranza Rubattu
- IRCCS Neuromed, Pozzilli, Italy
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Mariangela Peruzzi
- Mediterranea Cardiocentro, Napoli, Italy
- Department of Clinical, Internal Medicine, Anaesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Francesco Versaci
- Haemodynamic and Cardiology Unit, "Santa Maria Goretti" Hospital, Latina, Italy
- Department of System Medicine, "Tor Vergata" University, Rome, Italy
| | - Ernesto Greco
- Department of Clinical, Internal Medicine, Anaesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Antonella Calogero
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Elena De Falco
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Mediterranea Cardiocentro, Napoli, Italy
| | - Giacomo Frati
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Sebastiano Sciarretta
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
33
|
Penna C, Comità S, Tullio F, Alloatti G, Pagliaro P. Challenges facing the clinical translation of cardioprotection: 35 years after the discovery of ischemic preconditioning. Vascul Pharmacol 2022; 144:106995. [DOI: 10.1016/j.vph.2022.106995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/17/2022] [Accepted: 04/16/2022] [Indexed: 12/19/2022]
|
34
|
Forte E, Ramialison M, Nim HT, Mara M, Li JY, Cohn R, Daigle SL, Boyd S, Stanley EG, Elefanty AG, Hinson JT, Costa MW, Rosenthal NA, Furtado MB. Adult mouse fibroblasts retain organ-specific transcriptomic identity. eLife 2022; 11:71008. [PMID: 35293863 PMCID: PMC8959603 DOI: 10.7554/elife.71008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 03/15/2022] [Indexed: 01/18/2023] Open
Abstract
Organ fibroblasts are essential components of homeostatic and diseased tissues. They participate in sculpting the extracellular matrix, sensing the microenvironment, and communicating with other resident cells. Recent studies have revealed transcriptomic heterogeneity among fibroblasts within and between organs. To dissect the basis of interorgan heterogeneity, we compare the gene expression of murine fibroblasts from different tissues (tail, skin, lung, liver, heart, kidney, and gonads) and show that they display distinct positional and organ-specific transcriptome signatures that reflect their embryonic origins. We demonstrate that expression of genes typically attributed to the surrounding parenchyma by fibroblasts is established in embryonic development and largely maintained in culture, bioengineered tissues and ectopic transplants. Targeted knockdown of key organ-specific transcription factors affects fibroblast functions, in particular genes involved in the modulation of fibrosis and inflammation. In conclusion, our data reveal that adult fibroblasts maintain an embryonic gene expression signature inherited from their organ of origin, thereby increasing our understanding of adult fibroblast heterogeneity. The knowledge of this tissue-specific gene signature may assist in targeting fibrotic diseases in a more precise, organ-specific manner.
Collapse
Affiliation(s)
| | - Mirana Ramialison
- Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
| | - Hieu T Nim
- Faculty of Information Technology, Monash University, Clayton, Australia
| | | | - Jacky Y Li
- Murdoch Children's Research Institute, Parkville, Australia
| | - Rachel Cohn
- Jackson Laboratory, Farmington, United States
| | | | - Sarah Boyd
- Centre for Inflammatory Diseases, Monash University, Clayton, Australia
| | | | | | | | | | | | | |
Collapse
|
35
|
Arslan U, Moruzzi A, Nowacka J, Mummery CL, Eckardt D, Loskill P, Orlova VV. Microphysiological stem cell models of the human heart. Mater Today Bio 2022; 14:100259. [PMID: 35514437 PMCID: PMC9062349 DOI: 10.1016/j.mtbio.2022.100259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 11/10/2022] Open
Abstract
Models of heart disease and drug responses are increasingly based on human pluripotent stem cells (hPSCs) since their ability to capture human heart (dys-)function is often better than animal models. Simple monolayer cultures of hPSC-derived cardiomyocytes, however, have shortcomings. Some of these can be overcome using more complex, multi cell-type models in 3D. Here we review modalities that address this, describe efforts to tailor readouts and sensors for monitoring tissue- and cell physiology (exogenously and in situ) and discuss perspectives for implementation in industry and academia.
Collapse
Affiliation(s)
- Ulgu Arslan
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Alessia Moruzzi
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- Institute for Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Joanna Nowacka
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, the Netherlands
| | | | - Peter Loskill
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- Institute for Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
- 3R-Center for in Vitro Models and Alternatives to Animal Testing, Tübingen, Germany
| | - Valeria V. Orlova
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, the Netherlands
| |
Collapse
|
36
|
Cardiac regeneration following myocardial infarction: the need for regeneration and a review of cardiac stromal cell populations used for transplantation. Biochem Soc Trans 2022; 50:269-281. [PMID: 35129611 PMCID: PMC9042388 DOI: 10.1042/bst20210231] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/02/2022] [Accepted: 01/06/2022] [Indexed: 02/07/2023]
Abstract
Myocardial infarction is a leading cause of death globally due to the inability of the adult human heart to regenerate after injury. Cell therapy using cardiac-derived progenitor populations emerged about two decades ago with the aim of replacing cells lost after ischaemic injury. Despite early promise from rodent studies, administration of these populations has not translated to the clinic. We will discuss the need for cardiac regeneration and review the debate surrounding how cardiac progenitor populations exert a therapeutic effect following transplantation into the heart, including their ability to form de novo cardiomyocytes and the release of paracrine factors. We will also discuss limitations hindering the cell therapy field, which include the challenges of performing cell-based clinical trials and the low retention of administered cells, and how future research may overcome them.
Collapse
|
37
|
Abstract
The heart is a never-stopping engine that relies on a formidable pool of mitochondria to generate energy and propel pumping. Because dying cardiomyocytes cannot be replaced, this high metabolic rate creates the challenge of preserving organelle fitness and cell function for life. Here, we provide an immunologist's perspective on how the heart solves this challenge, which is in part by incorporating macrophages as an integral component of the myocardium. Cardiac macrophages surround cardiomyocytes and capture dysfunctional mitochondria that these cells eject to the milieu, effectively establishing a client cell-support cell interaction. We refer to this heterologous partnership as heterophagy. Notably, this process shares analogies with other biological systems, is essential for proteostasis and metabolic fitness of cardiomyocytes, and unveils a remarkable degree of dependence of the healthy heart on immune cells for everyday function.
Collapse
Affiliation(s)
- José A Nicolás-Ávila
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Spain (J.A.N.-A., L.P.-C., P.M.-C., A.H.)
| | - Laura Pena-Couso
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Spain (J.A.N.-A., L.P.-C., P.M.-C., A.H.)
| | - Pura Muñoz-Cánoves
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Spain (J.A.N.-A., L.P.-C., P.M.-C., A.H.).,Department of Experimental & Health Sciences, Universitat Pompeu Fabra, CIBERNED, Spain (P.M.-C.).,ICREA, Spain (P.M.-C.)
| | - Andrés Hidalgo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Spain (J.A.N.-A., L.P.-C., P.M.-C., A.H.)
| |
Collapse
|
38
|
Jayawardena E, Medzikovic L, Ruffenach G, Eghbali M. Role of miRNA-1 and miRNA-21 in Acute Myocardial Ischemia-Reperfusion Injury and Their Potential as Therapeutic Strategy. Int J Mol Sci 2022; 23:ijms23031512. [PMID: 35163436 PMCID: PMC8836257 DOI: 10.3390/ijms23031512] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
Coronary artery disease remains the leading cause of death. Acute myocardial infarction (MI) is characterized by decreased blood flow to the coronary arteries, resulting in cardiomyocytes death. The most effective strategy for treating an MI is early and rapid myocardial reperfusion, but restoring blood flow to the ischemic myocardium can induce further damage, known as ischemia-reperfusion (IR) injury. Novel therapeutic strategies are critical to limit myocardial IR injury and improve patient outcomes following reperfusion intervention. miRNAs are small non-coding RNA molecules that have been implicated in attenuating IR injury pathology in pre-clinical rodent models. In this review, we discuss the role of miR-1 and miR-21 in regulating myocardial apoptosis in ischemia-reperfusion injury in the whole heart as well as in different cardiac cell types with special emphasis on cardiomyocytes, fibroblasts, and immune cells. We also examine therapeutic potential of miR-1 and miR-21 in preclinical studies. More research is necessary to understand the cell-specific molecular principles of miRNAs in cardioprotection and application to acute myocardial IR injury.
Collapse
|
39
|
Chimenti I, Sattler S, del Monte-Nieto G, Forte E. Editorial: Fibrosis and Inflammation in Tissue Pathophysiology. Front Physiol 2022; 12:830683. [PMID: 35126187 PMCID: PMC8814660 DOI: 10.3389/fphys.2021.830683] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/29/2021] [Indexed: 12/16/2022] Open
Affiliation(s)
- Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
- Mediterranea Cardiocentro, Naples, Italy
- *Correspondence: Isotta Chimenti
| | - Susanne Sattler
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | - Elvira Forte
- The Jackson Laboratory, Bar Harbor, ME, United States
| |
Collapse
|
40
|
Intrauterine inflammation exacerbates maladaptive remodeling of the immature myocardium after preterm birth in lambs. Pediatr Res 2022; 92:1555-1565. [PMID: 35277596 PMCID: PMC9771797 DOI: 10.1038/s41390-022-01955-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/14/2021] [Accepted: 12/14/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Antenatal conditions that are linked with preterm birth, such as intrauterine inflammation, can influence fetal cardiac development thereby rendering the heart more vulnerable to the effects of prematurity. We aimed to investigate the effect of intrauterine inflammation, consequent to lipopolysaccharide exposure, on postnatal cardiac growth and maturation in preterm lambs. METHODS Preterm lambs (~129 days gestational age) exposed antenatally to lipopolysaccharide or saline were managed according to contemporary neonatal care and studied at postnatal day 7. Age-matched fetal controls were studied at ~136 days gestational age. Cardiac tissue was sampled for molecular analyses and assessment of cardiac structure and cardiomyocyte maturation. RESULTS Lambs delivered preterm showed distinct ventricular differences in cardiomyocyte growth and maturation trajectories as well as remodeling of the left ventricular myocardium compared to fetal controls. Antenatal exposure to lipopolysaccharide resulted in further collagen deposition in the left ventricle and a greater presence of immune cells in the preterm heart. CONCLUSIONS Adverse impacts of preterm birth on cardiac structure and cardiomyocyte growth kinetics within the first week of postnatal life are exacerbated by intrauterine inflammation. The maladaptive remodeling of the cardiac structure and perturbed cardiomyocyte growth likely contribute to the increased vulnerability to cardiac dysfunction following preterm birth. IMPACT Preterm birth induces maladaptive cardiac remodeling and adversely impacts cardiomyocyte growth kinetics within the first week of life in sheep. These effects of prematurity on the heart are exacerbated when preterm birth is preceded by exposure to intrauterine inflammation, a common antecedent of preterm birth. Inflammatory injury to the fetal heart coupled with preterm birth consequently alters neonatal cardiac growth and maturation and thus, may potentially influence long-term cardiac function and health.
Collapse
|
41
|
Buja LM, Mitchell RN. Basic pathobiology of cell-based therapies and cardiac regenerative medicine. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00016-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
42
|
Tombor LS, Dimmeler S. Why is endothelial resilience key to maintain cardiac health? Basic Res Cardiol 2022; 117:35. [PMID: 35834003 PMCID: PMC9283358 DOI: 10.1007/s00395-022-00941-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 02/01/2023]
Abstract
Myocardial injury as induced by myocardial infarction results in tissue ischemia, which critically incepts cardiomyocyte death. Endothelial cells play a crucial role in restoring oxygen and nutrient supply to the heart. Latest advances in single-cell multi-omics, together with genetic lineage tracing, reveal a transcriptional and phenotypical adaptation to the injured microenvironment, which includes alterations in metabolic, mesenchymal, hematopoietic and pro-inflammatory signatures. The extent of transition in mesenchymal or hematopoietic cell lineages is still debated, but it is clear that several of the adaptive phenotypical changes are transient and endothelial cells revert back to a naïve cell state after resolution of injury responses. This resilience of endothelial cells to acute stress responses is important for preventing chronic dysfunction. Here, we summarize how endothelial cells adjust to injury and how this dynamic response contributes to repair and regeneration. We will highlight intrinsic and microenvironmental factors that contribute to endothelial cell resilience and may be targetable to maintain a functionally active, healthy microcirculation.
Collapse
Affiliation(s)
- Lukas S. Tombor
- Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany ,Faculty for Biological Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany ,Faculty for Biological Sciences, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
43
|
From Spheroids to Organoids: The Next Generation of Model Systems of Human Cardiac Regeneration in a Dish. Int J Mol Sci 2021; 22:ijms222413180. [PMID: 34947977 PMCID: PMC8708686 DOI: 10.3390/ijms222413180] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/02/2021] [Accepted: 12/05/2021] [Indexed: 12/12/2022] Open
Abstract
Organoids are tiny, self-organized, three-dimensional tissue cultures that are derived from the differentiation of stem cells. The growing interest in the use of organoids arises from their ability to mimic the biology and physiology of specific tissue structures in vitro. Organoids indeed represent promising systems for the in vitro modeling of tissue morphogenesis and organogenesis, regenerative medicine and tissue engineering, drug therapy testing, toxicology screening, and disease modeling. Although 2D cell cultures have been used for more than 50 years, even for their simplicity and low-cost maintenance, recent years have witnessed a steep rise in the availability of organoid model systems. Exploiting the ability of cells to re-aggregate and reconstruct the original architecture of an organ makes it possible to overcome many limitations of 2D cell culture systems. In vitro replication of the cellular micro-environment of a specific tissue leads to reproducing the molecular, biochemical, and biomechanical mechanisms that directly influence cell behavior and fate within that specific tissue. Lineage-specific self-organizing organoids have now been generated for many organs. Currently, growing cardiac organoid (cardioids) from pluripotent stem cells and cardiac stem/progenitor cells remains an open challenge due to the complexity of the spreading, differentiation, and migration of cardiac muscle and vascular layers. Here, we summarize the evolution of biological model systems from the generation of 2D spheroids to 3D organoids by focusing on the generation of cardioids based on the currently available laboratory technologies and outline their high potential for cardiovascular research.
Collapse
|
44
|
Fischesser DM, Bo B, Benton RP, Su H, Jahanpanah N, Haworth KJ. Controlling Reperfusion Injury With Controlled Reperfusion: Historical Perspectives and New Paradigms. J Cardiovasc Pharmacol Ther 2021; 26:504-523. [PMID: 34534022 DOI: 10.1177/10742484211046674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cardiac reperfusion injury is a well-established outcome following treatment of acute myocardial infarction and other types of ischemic heart conditions. Numerous cardioprotection protocols and therapies have been pursued with success in pre-clinical models. Unfortunately, there has been lack of successful large-scale clinical translation, perhaps in part due to the multiple pathways that reperfusion can contribute to cell death. The search continues for new cardioprotection protocols based on what has been learned from past results. One class of cardioprotection protocols that remain under active investigation is that of controlled reperfusion. This class consists of those approaches that modify, in a controlled manner, the content of the reperfusate or the mechanical properties of the reperfusate (e.g., pressure and flow). This review article first provides a basic overview of the primary pathways to cell death that have the potential to be addressed by various forms of controlled reperfusion, including no-reflow phenomenon, ion imbalances (particularly calcium overload), and oxidative stress. Descriptions of various controlled reperfusion approaches are described, along with summaries of both mechanistic and outcome-oriented studies at the pre-clinical and clinical phases. This review will constrain itself to approaches that modify endogenously-occurring blood components. These approaches include ischemic postconditioning, gentle reperfusion, controlled hypoxic reperfusion, controlled hyperoxic reperfusion, controlled acidotic reperfusion, and controlled ionic reperfusion. This review concludes with a discussion of the limitations of past approaches and how they point to potential directions of investigation for the future.
Collapse
Affiliation(s)
- Demetria M Fischesser
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, 2514University of Cincinnati, Cincinnati, OH, USA
| | - Bin Bo
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, 2514University of Cincinnati, Cincinnati, OH, USA
| | - Rachel P Benton
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, 2514University of Cincinnati, Cincinnati, OH, USA
| | - Haili Su
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, 2514University of Cincinnati, Cincinnati, OH, USA
| | - Newsha Jahanpanah
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, 2514University of Cincinnati, Cincinnati, OH, USA
| | - Kevin J Haworth
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, 2514University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
45
|
Veltman D, Wu M, Pokreisz P, Claus P, Gillijns H, Caluwé E, Vanhaverbeke M, Gsell W, Himmelreich U, Sinnaeve PR, Janssens SP. Clec4e-Receptor Signaling in Myocardial Repair After Ischemia-Reperfusion Injury. JACC Basic Transl Sci 2021; 6:631-646. [PMID: 34466750 PMCID: PMC8385568 DOI: 10.1016/j.jacbts.2021.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/02/2021] [Accepted: 07/02/2021] [Indexed: 01/02/2023]
Abstract
The role of the CLEC4E during myocardial healing after ischemia-reperfusion injury is unknown. CLEC4E deletion is associated with reduced cardiac injury, inflammation, and left ventricular structural and functional remodeling. CLEC4E is a promising target to modulate myocardial inflammation and enhance repair after ischemia-reperfusion injury.
The bacterial C-type lectin domain family 4 member E (CLEC4E) has an important role in sterile inflammation, but its role in myocardial repair is unknown. Using complementary approaches in porcine, murine, and human samples, we show that CLEC4E expression levels in the myocardium and in blood correlate with the extent of myocardial injury and left ventricular (LV) functional impairment. CLEC4E expression is markedly increased in the vasculature, cardiac myocytes, and infiltrating leukocytes in the ischemic heart. Loss of Clec4e signaling is associated with reduced acute cardiac injury, neutrophil infiltration, and infarct size. Reduced myocardial injury in Clec4e–/– translates into significantly improved LV structural and functional remodeling at 4 weeks’ follow-up. The early transcriptome of LV tissue from Clec4e–/– mice versus wild-type mice reveals significant upregulation of transcripts involved in myocardial metabolism, radical scavenging, angiogenesis, and extracellular matrix organization. Therefore, targeting CLEC4E in the early phase of ischemia-reperfusion injury is a promising therapeutic strategy to modulate myocardial inflammation and enhance repair after ischemia-reperfusion injury.
Collapse
Key Words
- ACS, acute coronary syndrome
- AMI, acute myocardial infarction
- ANOVA, analysis of variance
- CAD, coronary artery disease
- CLEC4E
- CLEC4E, C-type lectin domain family 4 member E
- CMC, cardiac myocyte
- Car3, carbonic anhydrase 3
- Cxcl2, CXC chemokine ligand 2
- Cxcr2, CXC chemokine receptor 2
- DAMP, damage-associated molecular pattern
- ECM, extracellular matrix
- ESV, end-systolic volume
- Efna2, ephrin A2
- Grk2, G protein–coupled receptor kinase 2
- I/R, ischemia-reperfusion
- LAD, left anterior descending coronary artery
- LV, left ventricular
- MPO, myeloperoxidase
- MRI, magnetic resonance imaging
- NS, not significant
- PRR, pattern recognition receptor
- RNA, ribonucleic acid
- SMC, smooth muscle cell
- STEMI, ST-segment elevation myocardial infarction
- TnT, troponin T
- WT, wild-type
- hs-TnI, high-sensitivity troponin I
- inflammation
- ischemia-reperfusion injury
- magnetic resonance imaging
- myocardial remodeling
- qRT-PCR, quantitative reverse transcription polymerase chain reaction
Collapse
Affiliation(s)
- Denise Veltman
- Department of Cardiovascular Sciences, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Ming Wu
- Department of Cardiovascular Sciences, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Peter Pokreisz
- Department of Cardiovascular Sciences, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Piet Claus
- Department of Cardiovascular Sciences, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Hilde Gillijns
- Department of Cardiovascular Sciences, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Ellen Caluwé
- Department of Cardiovascular Sciences, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Maarten Vanhaverbeke
- Department of Cardiovascular Diseases, University Hospital Leuven, Leuven, Belgium
| | - Willy Gsell
- Department of Imaging and Pathology, Biomedical MRI, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Uwe Himmelreich
- Department of Imaging and Pathology, Biomedical MRI, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Peter R. Sinnaeve
- Department of Cardiovascular Sciences, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
- Department of Cardiovascular Diseases, University Hospital Leuven, Leuven, Belgium
| | - Stefan P. Janssens
- Department of Cardiovascular Sciences, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
- Department of Cardiovascular Diseases, University Hospital Leuven, Leuven, Belgium
- Address for correspondence: Dr Stefan P. Janssens, Department of Cardiovascular Sciences, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
46
|
New perspectives of the cardiac cellular landscape: mapping cellular mediators of cardiac fibrosis using single-cell transcriptomics. Biochem Soc Trans 2021; 48:2483-2493. [PMID: 33259583 DOI: 10.1042/bst20191255] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022]
Abstract
Single-cell transcriptomics enables inference of context-dependent phenotypes of individual cells and determination of cellular diversity of complex tissues. Cardiac fibrosis is a leading factor in the development of heart failure and a major cause of morbidity and mortality worldwide with no effective treatment. Single-cell RNA-sequencing (scRNA-seq) offers a promising new platform to identify new cellular and molecular protagonists that may drive cardiac fibrosis and development of heart failure. This review will summarize the application scRNA-seq for understanding cardiac fibrosis and development of heart failure. We will also discuss some key considerations in interpreting scRNA-seq data and some of its limitations.
Collapse
|
47
|
Li X, Li L, Lei W, Chua HZ, Li Z, Huang X, Wang Q, Li N, Zhang H. Traditional Chinese medicine as a therapeutic option for cardiac fibrosis: Pharmacology and mechanisms. Biomed Pharmacother 2021; 142:111979. [PMID: 34358754 DOI: 10.1016/j.biopha.2021.111979] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/05/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases are one of the leading causes of death worldwide and cardiac fibrosis is a common pathological process for cardiac remodeling in cardiovascular diseases. Cardiac fibrosis not only accelerates the deterioration progress of diseases but also becomes a pivotal contributor for futile treatment in clinical cardiovascular trials. Although cardiac fibrosis is common and prevalent, effective medicines to provide sufficient clinical intervention for cardiac fibrosis are still unavailable. Traditional Chinese medicine (TCM) is the natural essence experienced boiling, fry, and other processing methods, including active ingredients, extracts, and herbal formulas, which have been applied to treat human diseases for a long history. Recently, research has increasingly focused on the great potential of TCM for the prevention and treatment of cardiac fibrosis. Here, we aim to clarify the identified pro-fibrotic mechanisms and intensively summarize the application of TCM in improving cardiac fibrosis by working on these mechanisms. Through comprehensively analyzing, TCM mainly regulates the following pathways during ameliorating cardiac fibrosis: attenuation of inflammation and oxidative stress, inhibition of cardiac fibroblasts activation, reduction of extracellular matrix accumulation, modulation of the renin-angiotensin-aldosterone system, modulation of autophagy, regulation of metabolic-dependent mechanisms, and targeting microRNAs. We also discussed the deficiencies and the development direction of anti-fibrotic therapies on cardiac fibrosis. The data reviewed here demonstrates that TCM shows a robust effect on alleviating cardiac fibrosis, which provides us a rich source of new drugs or drug candidates. Besides, we also hope this review may give some enlightenment for treating cardiac fibrosis in clinical practice.
Collapse
Affiliation(s)
- Xiao Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Lin Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Wei Lei
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Hui Zi Chua
- Evidence-Based Medicine Center, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Zining Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Xianglong Huang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China.
| | - Qilong Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Nan Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Han Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
48
|
Park Y, Koh JS, Lee JH, Park JH, Shin ES, Oh JH, Chun W, Lee SY, Bae JW, Kim JS, Kim W, Suh JW, Yang DH, Hong YJ, Chan MY, Kang MG, Park HW, Hwang SJ, Hwang JY, Ahn JH, Choi SW, Jeong YH. Effect of Ticagrelor on Left Ventricular Remodeling in Patients With ST-Segment Elevation Myocardial Infarction (HEALING-AMI). JACC Cardiovasc Interv 2021; 13:2220-2234. [PMID: 33032710 DOI: 10.1016/j.jcin.2020.08.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/17/2020] [Accepted: 08/04/2020] [Indexed: 12/30/2022]
Abstract
OBJECTIVES The aim of this study was to evaluate the effect of ticagrelor versus clopidogrel on left ventricular (LV) remodeling after reperfusion of ST-segment elevation myocardial infarction (STEMI) in humans. BACKGROUND Animal studies have demonstrated that ticagrelor compared with clopidogrel better protects myocardium against reperfusion injury and improves remodeling after myocardial infarction. METHODS In this investigator-initiated, randomized, open-label, assessor-blinded trial performed at 10 centers in Korea, patients were enrolled if they had naive STEMI successfully treated with primary percutaneous coronary intervention (PCI) and at least 6-month planned duration of dual-antiplatelet treatment. The coprimary endpoints were LV remodeling index (LVRI) (a relative change of LV end-diastolic volume) measured on 3-dimensional echocardiography and N-terminal pro-B-type natriuretic peptide level at 6 months. RESULTS Among initially enrolled patients with STEMI (n = 336), 139 in each group completed the study. LVRI at 6 months was numerically lower with ticagrelor versus clopidogrel (0.6 ± 18.6% vs. 4.5 ± 16.5%; p = 0.095). Ticagrelor significantly reduced the 6-month level of N-terminal pro-B-type natriuretic peptide (173 ± 141 pg/ml vs. 289 ± 585 pg/ml; p = 0.028). These differences were prominent in patients with pre-PCI TIMI (Thrombolysis In Myocardial Infarction) flow grade 0. By multivariate analysis, ticagrelor versus clopidogrel reduced the risk for positive LV remodeling (LVRI >0%) (odds ratio: 0.56; 95% confidence interval: 0.33 to 0.95; p = 0.030). The LV end-diastolic volume index remained unchanged during ticagrelor treatment (from 54.7 ± 12.2 to 54.2 ± 12.2 ml/m2; p = 0.629), but this value increased over time during clopidogrel treatment (from 54.6 ± 11.3 to 56.4 ± 13.9 ml/m2; p = 0.056) (difference -2.3 ml/m2; 95% confidence interval: -4.8 to 0.2 ml/m2; p = 0.073). Ticagrelor reduced LV end-systolic volume index (from 27.0 ± 8.5 to 24.7 ± 8.4 ml/m2; p < 0.001), whereas no reduction was seen with clopidogrel (from 26.2 ± 8.9 to 25.6 ± 11.0 ml/m2; p = 0.366) (difference -1.8 ml/m2; 95% confidence interval: -3.5 to -0.1 ml/m2; p = 0.040). CONCLUSIONS Ticagrelor was superior to clopidogrel for LV remodeling after reperfusion of STEMI with primary PCI. (High Platelet Inhibition With Ticagrelor to Improve Left Ventricular Remodeling in Patients With ST Segment Elevation Myocardial Infarction [HEALING-AMI]; NCT02224534).
Collapse
Affiliation(s)
- Yongwhi Park
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon, South Korea
| | - Jin Sin Koh
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, South Korea
| | - Jae-Hwan Lee
- Department of Cardiology, Chungnam National University Hospital, Daejeon, South Korea
| | - Jae-Hyeong Park
- Department of Cardiology, Chungnam National University Hospital, Daejeon, South Korea
| | - Eun-Seok Shin
- Department of Cardiology, Ulsan Medical Center, Ulsan Hospital, Ulsan, South Korea
| | - Ju Hyeon Oh
- Division of Cardiology, Department of Internal Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, South Korea
| | - Woojung Chun
- Division of Cardiology, Department of Internal Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, South Korea
| | - Sang Yeub Lee
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, South Korea
| | - Jang-Whan Bae
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, South Korea
| | - Jeong Su Kim
- Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Weon Kim
- Cardiovascular Department of Internal Medicine, Kyung Hee University Hospital, Seoul, South Korea
| | - Jung-Won Suh
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Dong Heon Yang
- Department of Cardiology, Kyungpook National University Hospital, Daegu, South Korea
| | - Young-Joon Hong
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, South Korea
| | - Mark Y Chan
- Singapore National University Heart Center, Singapore National University Hospital, Singapore, Singapore
| | - Min Gyu Kang
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, South Korea
| | - Hyun-Woong Park
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, South Korea
| | - Seok-Jae Hwang
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, South Korea
| | - Jin-Yong Hwang
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, South Korea
| | - Jong-Hwa Ahn
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon, South Korea
| | - Si Wan Choi
- Department of Cardiology, Chungnam National University Hospital, Daejeon, South Korea.
| | - Young-Hoon Jeong
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon, South Korea.
| |
Collapse
|
49
|
Viola M, de Jager SCA, Sluijter JPG. Targeting Inflammation after Myocardial Infarction: A Therapeutic Opportunity for Extracellular Vesicles? Int J Mol Sci 2021; 22:ijms22157831. [PMID: 34360595 PMCID: PMC8346058 DOI: 10.3390/ijms22157831] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/15/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
After myocardial infarction (MI), a strong inflammatory response takes place in the heart to remove the dead tissue resulting from ischemic injury. A growing body of evidence suggests that timely resolution of this inflammatory process may aid in the prevention of adverse cardiac remodeling and heart failure post-MI. The present challenge is to find a way to stimulate this process without interfering with the reparative role of the immune system. Extracellular vesicles (EVs) are natural membrane particles that are released by cells and carry different macromolecules, including proteins and non-coding RNAs. In recent years, EVs derived from various stem and progenitor cells have been demonstrated to possess regenerative properties. They can provide cardioprotection via several mechanisms of action, including immunomodulation. In this review, we summarize the role of the innate immune system in post-MI healing. We then discuss the mechanisms by which EVs modulate cardiac inflammation in preclinical models of myocardial injury through regulation of monocyte influx and macrophage function. Finally, we provide suggestions for further optimization of EV-based therapy to improve its potential for the treatment of MI.
Collapse
Affiliation(s)
- Margarida Viola
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands;
- UMC Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, 3584 CS Utrecht, The Netherlands
| | - Saskia C. A. de Jager
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands;
- UMC Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, 3584 CS Utrecht, The Netherlands
- Correspondence: (S.C.A.d.J.); (J.P.G.S.)
| | - Joost P. G. Sluijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands;
- UMC Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, 3584 CS Utrecht, The Netherlands
- Correspondence: (S.C.A.d.J.); (J.P.G.S.)
| |
Collapse
|
50
|
Lai Y, Zhou X, Guo F, Jin X, Meng G, Zhou L, Chen H, Liu Z, Yu L, Jiang H. Non-invasive transcutaneous vagal nerve stimulation improves myocardial performance in doxorubicin-induced cardiotoxicity. Cardiovasc Res 2021; 118:1821-1834. [PMID: 34145895 DOI: 10.1093/cvr/cvab209] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 06/17/2021] [Indexed: 12/29/2022] Open
Abstract
AIMS The clinical use of antitumor agent doxorubicin (DOX) is hampered by its dose-dependent cardiotoxicity. Development of highly efficient and safe adjuvant intervention for preventing DOX-induced adverse cardiac events is urgently needed. We aimed to investigate whether transcutaneous vagal nerve stimulation (tVNS) plays a cardio-protective role in DOX-induced cardiotoxicity. METHODS AND RESULTS Healthy male adult Sprague Dawley rats were used in the experiment and were randomly divided into four groups including control, DOX, tVNS and DOX+tVNS groups. A cumulative dose of 15 mg/kg DOX was intraperitoneally injected into rats to generate cardiotoxicity. Non-invasive tVNS was conducted for 6 weeks (30 min/day). After six-week intervention, the indices from the echocardiography revealed that tVNS significantly improved left ventricular function compared to the DOX group. The increased malondialdehyde (MDA) and Interleukin-1β (IL-1β), and decreased superoxide dismutase (SOD) were observed in the DOX group, while tVNS significantly prevented these changes. From cardiac histopathological analysis, the DOX+tVNS group showed a mild myocardial damage, and decreases in cardiac fibrosis and myocardial apoptosis compared to the DOX group. Heart rate variability (HRV) analysis showed that tVNS significantly inhibited DOX-induced sympathetic hyperactivity compared to the DOX group. Additionally, the results of RNA-sequencing analysis showed that there were 245 differentially expressed genes in the DOX group compared to the control group, among which 39 genes were downregulated by tVNS and most of these genes were involved in immune system. Moreover, tVNS significantly downregulated the relative mRNA expressions of chemokine-related genes and macrophages recruitment compared to the DOX group. CONCLUSION These results suggest that tVNS prevented DOX-induced cardiotoxicity by rebalancing autonomic tone, ameliorating cardiac dysfunction and remodeling. Notably, crosstalk between autonomic neuromodulation and innate immune cells macrophages mediated by chemokines might be involved in the underlying mechanisms. A TRANSLATIONAL PERSPECTIVE Non-invasive tVNS has been identified an effective neuromodulation strategy exerting beneficial effects on rebalancing autonomic tone and cardiac pathological conditions. The present study provided direct evidence for a beneficial role of tVNS in preventing DOX-induced autonomic dysfunction and cardiotoxicity in vivo. Additionally, recent studies revealed the importance of sympathetic nerve fibers involving in tumorigenesis and the benefits of higher vagal tone for tumor prognosis either in animal or human trials. Together, tVNS may not only become a novel, nonpharmacological adjuvant therapy for preventing doxorubicin-induced cardiotoxicity, but also may be beneficial for prognosis of cancer patients during chemotherapy. In our future study, we would investigate the effect of tVNS on both combined chemotherapy-induced cardiotoxicity and the antitumor efficacy of DOX in tumor models.
Collapse
Affiliation(s)
- Yanqiu Lai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| | - Xiaoya Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| | - Fuding Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| | - Xiaoxing Jin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| | - Guannan Meng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| | - Liping Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| | - Hu Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| | - Zhihao Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| | - Lilei Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| |
Collapse
|