1
|
Shooshtarian AK, O'Gallagher K, Shah AM, Zhang M. SERCA2a dysfunction in the pathophysiology of heart failure with preserved ejection fraction: a direct role is yet to be established. Heart Fail Rev 2025; 30:545-564. [PMID: 39843817 PMCID: PMC11991975 DOI: 10.1007/s10741-025-10487-1] [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: 01/10/2025] [Indexed: 01/24/2025]
Abstract
With rising incidence, mortality and limited therapeutic options, heart failure with preserved ejection fraction (HFpEF) remains one of the most important topics in cardiovascular medicine today. Characterised by left ventricular diastolic dysfunction partially due to impaired Ca2+ homeostasis, one ion channel in particular, SarcoEndoplasmic Reticulum Ca2+-ATPase (SERCA2a), may play a significant role in its pathophysiology. A better understanding of the complex mechanisms interplaying to contribute to SERCA2a dysfunction will help develop treatments targeting it and thus address the growing clinical challenge HFpEF poses. This review examines the conflicting evidence present for changes in SERCA2a expression and activity in HFpEF, explores potential underlying mechanisms, and finally evaluates the drug and gene therapy trials targeting SERCA2a in heart failure. Recent positive results from trials involving widely used anti-diabetic agents such as sodium-glucose co-transporter protein 2 inhibitors (SGLT2i) and glucagon-like peptide-1 (GLP-1) agonists offer advancement in HFpEF management. The potential interplay between these agents and SERCA2a regulation presents a novel angle that could open new avenues for modulating diastolic function; however, the mechanistic research in this emerging field is limited. Overall, the direct role of SERCA2a dysfunction in HFpEF remains undetermined, highlighting the need for well-designed pre-clinical studies and robust clinical trials.
Collapse
Affiliation(s)
- Adam Kia Shooshtarian
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Research Excellence, London, UK
| | - Kevin O'Gallagher
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Research Excellence, London, UK
| | - Ajay M Shah
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Research Excellence, London, UK
| | - Min Zhang
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Research Excellence, London, UK.
| |
Collapse
|
2
|
Landim-Vieira M, Nieto Morales PF, ElSafty S, Kahmini AR, Ranek MJ, Solís C. The role of mechanosignaling in the control of myocardial mass. Am J Physiol Heart Circ Physiol 2025; 328:H622-H638. [PMID: 39739566 DOI: 10.1152/ajpheart.00277.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 12/03/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025]
Abstract
Regulation of myocardial mass is key for maintaining cardiovascular health. This review highlights the complex and regulatory relationship between mechanosignaling and myocardial mass, influenced by many internal and external factors including hemodynamic and microgravity, respectively. The heart is a dynamic organ constantly adapting to changes in workload (preload and afterload) and mechanical stress exerted on the myocardium, influencing both physiological adaptations and pathological remodeling. Mechanosignaling pathways, such as the mitogen-activated protein kinases (MAPKs) and the phosphoinositide 3-kinases and serine/threonine kinase (PI3K/Akt) pathways, mediate downstream effects on gene expression and play key roles in transducing mechanical cues into biochemical signals, thereby modulating cellular processes, including control of myocardial mass. Dysregulation of these processes can lead to pathological cardiac remodeling, such as hypertrophic cardiomyopathy. Furthermore, recent studies have highlighted the importance of protein quality control mechanisms, such as the ubiquitin-proteasome system, in settings of extreme physiological conditions that alter the heart workload such as pregnancy and microgravity. Overall, this review provides a thorough insight into how mechanical signals are converted into chemical signals to regulate myocardial mass in both healthy and diseased conditions.
Collapse
Affiliation(s)
- Maicon Landim-Vieira
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Paula F Nieto Morales
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Summer ElSafty
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Aida Rahimi Kahmini
- Department of Health, Nutrition, and Food Science, Florida State University, Tallahassee, Florida, United States
| | - Mark J Ranek
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, Maryland, United States
| | - Christopher Solís
- Department of Health, Nutrition, and Food Science, Florida State University, Tallahassee, Florida, United States
| |
Collapse
|
3
|
Chu Z, Zhang Y, Zhang Y, Chen H, Zhang D, Hao Q, Wang Z, Sun M, Zhao X, Liu Y. A novel perspective on the regulation of cardiac cell beating: cardiac cell under mechanical stimulation acts as "cell activation button" to activate adjacent cardiac cell. J Nanobiotechnology 2025; 23:141. [PMID: 40001199 PMCID: PMC11863431 DOI: 10.1186/s12951-025-03244-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
The regulation of cardiac cell beating is of great significance for understanding cardiac coordination mechanisms and the treatment of cardiovascular diseases. Inspired by this natural "cell regulates cell" mode in which sinoatrial node cells regulate atrial myocytes, this study presented a novel method to replicate this behavior in vitro through mechanical stimulation. Primary cardiac cells from Sprague-Dawley rats were isolated, cultured in 2D substrates, and applied to precise mechanical stimulation by developing a micro-manipulation platform. We demonstrated that a mechanical probe can act as an external activation device for quiescent cardiac cells, transforming them into "activation cells" capable of activating adjacent "target cells" through bioelectrical coupling. Calcium imaging with Fluo-4 probes revealed that this "cell activates cell" mechanism relies on mechano-electric feedback and calcium-mediated signal propagation via cell junctions. Our findings provide a non-destructive strategy to regulate target cardiac cell, deepen insights into the mechanical modulation of intercellular communication, and offer a framework for studying arrhythmias linked to abnormal cell-cell communication. This work combined mechanical intervention with biological signaling, advancing potential applications in cardiovascular therapeutics.
Collapse
Affiliation(s)
- Zhaotong Chu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Tianjin Key Laboratory of Intelligent Robotic (tjKLIR), Institute of Robotics and Automatic Information System (IRAIS), Nankai University, Tianjin, 300350, China
- Institute of Intelligence Technology and Robotic Systems, Shenzhen Research Institute of Nankai University, Shenzhen, 518083, China
| | - Yujie Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Tianjin Key Laboratory of Intelligent Robotic (tjKLIR), Institute of Robotics and Automatic Information System (IRAIS), Nankai University, Tianjin, 300350, China
- Institute of Intelligence Technology and Robotic Systems, Shenzhen Research Institute of Nankai University, Shenzhen, 518083, China
| | - Yidi Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Tianjin Key Laboratory of Intelligent Robotic (tjKLIR), Institute of Robotics and Automatic Information System (IRAIS), Nankai University, Tianjin, 300350, China
- Institute of Intelligence Technology and Robotic Systems, Shenzhen Research Institute of Nankai University, Shenzhen, 518083, China
| | - Hao Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Tianjin Key Laboratory of Intelligent Robotic (tjKLIR), Institute of Robotics and Automatic Information System (IRAIS), Nankai University, Tianjin, 300350, China
| | - Detian Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Tianjin Key Laboratory of Intelligent Robotic (tjKLIR), Institute of Robotics and Automatic Information System (IRAIS), Nankai University, Tianjin, 300350, China
| | - Qingzheng Hao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Tianjin Key Laboratory of Intelligent Robotic (tjKLIR), Institute of Robotics and Automatic Information System (IRAIS), Nankai University, Tianjin, 300350, China
| | - Zuqi Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Tianjin Key Laboratory of Intelligent Robotic (tjKLIR), Institute of Robotics and Automatic Information System (IRAIS), Nankai University, Tianjin, 300350, China
| | - Mingzhu Sun
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Tianjin Key Laboratory of Intelligent Robotic (tjKLIR), Institute of Robotics and Automatic Information System (IRAIS), Nankai University, Tianjin, 300350, China
- Institute of Intelligence Technology and Robotic Systems, Shenzhen Research Institute of Nankai University, Shenzhen, 518083, China
| | - Xin Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Tianjin Key Laboratory of Intelligent Robotic (tjKLIR), Institute of Robotics and Automatic Information System (IRAIS), Nankai University, Tianjin, 300350, China
- Institute of Intelligence Technology and Robotic Systems, Shenzhen Research Institute of Nankai University, Shenzhen, 518083, China
| | - Yaowei Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Tianjin Key Laboratory of Intelligent Robotic (tjKLIR), Institute of Robotics and Automatic Information System (IRAIS), Nankai University, Tianjin, 300350, China.
- Institute of Intelligence Technology and Robotic Systems, Shenzhen Research Institute of Nankai University, Shenzhen, 518083, China.
| |
Collapse
|
4
|
Zhang F, Wang Y, Zhu J, Wang J, Li Q, Feng J, Liu M, Li K, Tan J, Luo R, Yang H, Hou Y, He F, Qin J, Ding C, Yang W. Region- and Cell-type-Resolved Multiomic Atlas of the Heart. Mol Cell Proteomics 2025; 24:100922. [PMID: 39921206 DOI: 10.1016/j.mcpro.2025.100922] [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: 08/27/2023] [Revised: 01/02/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025] Open
Abstract
The heart is a vital muscular organ in vertebrate animals, responsible for maintaining blood circulation through rhythmic contraction. Although previous studies have investigated the heart proteome, the full hierarchical molecular network at cell-type- and region-resolved level, illustrating the specialized roles and crosstalk among different cell-types and regions, remains unclear. Here, we presented an atlas of cell-type-resolved proteome for mouse heart and region-resolved proteome for both mouse and human hearts. In-depth proteomic analysis identified 11,794 proteins across four cell-types and 11,995 proteins across six regions of the mouse heart. To further illustrate protein expression patterns in both physiological and pathological conditions, we conducted proteomic analysis on human heart samples from four regions with dilated cardiomyopathy (DCM). We quantified 8201 proteins in DCM tissue and 8316 proteins in adjacent unaffected myocardium tissue across the four human heart regions. Notably, we found that the retinoic acid synthesis pathway was significantly enriched in the DCM-affected left ventricle, and functional experiments demonstrated that all-trans retinoic acid efficiently rescued Ang II-induced myocardial hypertrophy and transverse aorta constriction-induced heart failure. In conclusion, our datasets uncovered the functional features of different cell-types and their synergistic cooperation centered by cell-type-specific transcription factors (TFs) in different regions, while these TF-TG (target gene) axes were significantly altered in DCM. Additionally, all-trans retinoic acid was demonstrated to be an efficient treatment for heart failure. This work presented a panoramic heart proteome map, offering a valuable resource for future cardiovascular research.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China; State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pathology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Yunzhi Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiajun Zhu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jinxi Wang
- Laboratory of Molecular Cardiology, CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai, China
| | - Qiang Li
- Laboratory of Molecular Cardiology, CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai, China
| | - Jinwen Feng
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mingwei Liu
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Kai Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiliang Tan
- Laboratory of Molecular Cardiology, CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai, China
| | - Rongkui Luo
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huangtian Yang
- Laboratory of Molecular Cardiology, CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai, China
| | - Yingyong Hou
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fuchu He
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Jun Qin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Chen Ding
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Departments of Cancer Research Institute, Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Translational Biomedical Engineering, Urumqi, China.
| | - Wenjun Yang
- Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Li S, Yin W, Liu Y, Yang C, Zhai Z, Xie M, Ye Z, Song X. Anisotropic conductive scaffolds for post-infarction cardiac repair. Biomater Sci 2025; 13:542-567. [PMID: 39688676 DOI: 10.1039/d4bm01109k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Myocardial infarction (MI) remains one of the most common and lethal cardiovascular diseases (CVDs), leading to the deterioration of cardiac function due to myocardial cell necrosis and fibrous scar tissue formation. Myocardial infarction (MI) remains one of the most common and lethal cardiovascular diseases (CVDs), leading to the deterioration of cardiac function due to myocardial cell necrosis and fibrous scar tissue formation. After MI, the anisotropic structural properties of myocardial tissue are destroyed, and its mechanical and electrical microenvironment also undergoes a series of pathological changes, such as ventricular wall stiffness, abnormal contraction, conduction network disruption, and irregular electrical signal propagation, which may further induce myocardial remodeling and even lead to heart failure. Therefore, bionic reconstruction of the anisotropic structural-mechanical-electrical microenvironment of the infarct area is key to repairing damaged myocardium. This article first summarizes the pathological changes in muscle fibre structure and conductive microenvironment after cardiac injury, and focuses on the classification and preparation methods of anisotropic conductive materials. In addition, the effects of these anisotropic conductive materials on the behavior of cardiac resident cells after myocardial infarction, such as directional growth, maturation, proliferation and migration, and the differentiation fate of stem cells and the possible molecular mechanisms involved are summarized. The design strategies for anisotropic conductive scaffolds for myocardial repair in future clinical research are also discussed, with the aim of providing new insights for researchers in related fields.
Collapse
Affiliation(s)
- Shimin Li
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China.
| | - Wenming Yin
- Department of Neurology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China
| | - Yali Liu
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong 528000, China
| | - Chang Yang
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China.
| | - Zitong Zhai
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China.
| | - Mingxiang Xie
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China.
| | - Ziyi Ye
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China.
| | - Xiaoping Song
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510910, China.
- Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
6
|
Jahan N, Islam S, Sivasundaram K, Ota A, Naito M, Kuroda J, Watanabe H. Role of versican in extracellular matrix formation: analysis in 3D culture. Am J Physiol Cell Physiol 2025; 328:C245-C257. [PMID: 39656505 DOI: 10.1152/ajpcell.00495.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/28/2024] [Accepted: 11/20/2024] [Indexed: 01/01/2025]
Abstract
Three-dimensional (3-D) cell culture creates an environment that allows cells to grow and interact with the surrounding extracellular framework. Versican plays a pivotal role in forming the provisional matrix, but it is still unclear how this proteoglycan affects the formation of the extracellular matrix. Here, we established a 3-D culture system using fibrin gel, which enables a long-term culture up to a month. With this system, we characterized fibroblasts obtained from the newborn knock-in homozygotes, termed R/R, expressing a disintegrin and metalloproteinase with thrombospondin motif (ADAMTS)-resistant versican and wild-type mice. R/R fibroblasts showed higher levels of versican deposition than wild-type, demonstrating that the initial ADAMTS-cleavage site is involved in versican turnover. These fibroblasts exhibited faster proliferation and myofibroblastic differentiation, concomitant with higher levels of transforming growth factor β-signaling. R/R fibroblast culture had higher deposition levels of fibronectin, type I and V collagens, and fibrillin-1, especially at the late stages of culture. These results suggest that versican expressed by dermal fibroblasts facilitates the extracellular matrix formation, at least by affecting fibroblast behavior.NEW & NOTEWORTHY We established a 3-D-culture system useful for analyzing fibroblast behavior and matrix formation. The initial cleavage site by ADAMTSs in versican core protein is mainly involved in versican turnover. Accumulating versican facilitates fibroblast proliferation and myofibroblastic differentiation in an autocrine or paracrine manner. Accumulating versican promotes the deposition of fibronectin and collagens.
Collapse
Affiliation(s)
- Nushrat Jahan
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| | - Shamima Islam
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| | - Karnan Sivasundaram
- Department of Biochemistry, School of Medicine, Aichi Medical University, Nagakute, Japan
| | - Akinobu Ota
- Department of Biochemistry, School of Medicine, Aichi Medical University, Nagakute, Japan
| | - Munekazu Naito
- Department of Anatomy, School of Medicine, Aichi Medical University, Nagakute, Japan
| | - Junpei Kuroda
- Laboratory of Pattern Formation, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| |
Collapse
|
7
|
Watanabe T, Hatayama N, Guo M, Yuhara S, Shinoka T. Bridging the Gap: Advances and Challenges in Heart Regeneration from In Vitro to In Vivo Applications. Bioengineering (Basel) 2024; 11:954. [PMID: 39451329 PMCID: PMC11505552 DOI: 10.3390/bioengineering11100954] [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: 08/06/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 10/26/2024] Open
Abstract
Cardiovascular diseases, particularly ischemic heart disease, area leading cause of morbidity and mortality worldwide. Myocardial infarction (MI) results in extensive cardiomyocyte loss, inflammation, extracellular matrix (ECM) degradation, fibrosis, and ultimately, adverse ventricular remodeling associated with impaired heart function. While heart transplantation is the only definitive treatment for end-stage heart failure, donor organ scarcity necessitates the development of alternative therapies. In such cases, methods to promote endogenous tissue regeneration by stimulating growth factor secretion and vascular formation alone are insufficient. Techniques for the creation and transplantation of viable tissues are therefore highly sought after. Approaches to cardiac regeneration range from stem cell injections to epicardial patches and interposition grafts. While numerous preclinical trials have demonstrated the positive effects of tissue transplantation on vasculogenesis and functional recovery, long-term graft survival in large animal models is rare. Adequate vascularization is essential for the survival of transplanted tissues, yet pre-formed microvasculature often fails to achieve sufficient engraftment. Recent studies report success in enhancing cell survival rates in vitro via tissue perfusion. However, the transition of these techniques to in vivo models remains challenging, especially in large animals. This review aims to highlight the evolution of cardiac patch and stem cell therapies for the treatment of cardiovascular disease, identify discrepancies between in vitro and in vivo studies, and discuss critical factors for establishing effective myocardial tissue regeneration in vivo.
Collapse
Affiliation(s)
- Tatsuya Watanabe
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (T.W.); (M.G.); (S.Y.)
| | - Naoyuki Hatayama
- Department of Anatomy, Aichi Medical University, Nagakute 480-1195, Japan;
| | - Marissa Guo
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (T.W.); (M.G.); (S.Y.)
- Department of Surgery, Ohio State University, Columbus, OH 43210, USA
| | - Satoshi Yuhara
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (T.W.); (M.G.); (S.Y.)
| | - Toshiharu Shinoka
- Department of Surgery, Ohio State University, Columbus, OH 43210, USA
- Department of Cardiothoracic Surgery, The Heart Center, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| |
Collapse
|
8
|
Raz M, Milo T, Glass DS, Mayo A, Alon U. Endocrine gland size is proportional to its target tissue size. iScience 2024; 27:110625. [PMID: 39224518 PMCID: PMC11367476 DOI: 10.1016/j.isci.2024.110625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 06/26/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Endocrine glands secrete hormones into the circulation to target distant tissues and regulate their functions. The qualitative relationship between hormone-secreting organs and their target tissues is well established, but a quantitative approach is currently limited. Quantification is important, as it could allow us to study the endocrine system using engineering concepts of optimality and tradeoffs. In this study, we collected literature data on 24 human hormones secreted from dedicated endocrine cells. We find that the number of endocrine cells secreting a hormone is proportional to the number of its target cells. A single endocrine cell serves approximately 2,000 target cells, a relationship that spans 6 orders of magnitude of cell numbers. This suggests an economic principle of cells working near their maximal capacity, and glands that are no bigger than they need to be.
Collapse
Affiliation(s)
- Moriya Raz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Tomer Milo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - David S. Glass
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Avi Mayo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Uri Alon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
9
|
Ketabat F, Alcorn J, Kelly ME, Badea I, Chen X. Cardiac Tissue Engineering: A Journey from Scaffold Fabrication to In Vitro Characterization. SMALL SCIENCE 2024; 4:2400079. [PMID: 40212070 PMCID: PMC11935279 DOI: 10.1002/smsc.202400079] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/18/2024] [Indexed: 04/13/2025] Open
Abstract
Cardiac tissue engineering has been rapidly evolving with diverse applications, ranging from the repair of fibrotic tissue caused by "adverse remodeling," to the replacement of specific segments of heart tissue, and ultimately to the creation of a whole heart. The repair or replacement of cardiac tissue often involves the development of tissue scaffolds or constructs and the subsequent assessment of their performance and functionality. For this, the design and/or selection of biomaterials, and cell types, scaffold fabrication, and in vitro characterizations are the first starting points, yet critical, to ensure success in subsequent implantation in vivo. This highlights the importance of scaffold fabrication and in vitro experiments/characterization with protocols for cardiac tissue engineering. Yet, a comprehensive and critical review of these has not been established and documented. As inspired, herein, the latest development and advances in scaffold fabrication and in vitro characterization for cardiac tissue engineering are critically reviewed, with focus on biomaterials, cell types, additive manufacturing techniques for scaffold fabrication, and common in vitro characterization techniques or methods. This article would be of benefit to the ones who are working on cardiac tissue engineering by providing insights into the scaffold fabrication and in vitro investigations.
Collapse
Affiliation(s)
- Farinaz Ketabat
- Division of Biomedical EngineeringUniversity of Saskatchewan57 Campus DriveSaskatoonS7N 5A9Canada
| | - Jane Alcorn
- College of Pharmacy and NutritionUniversity of Saskatchewan107 Wiggins RoadSaskatoonS7N 5E5SaskatchewanCanada
| | - Michael E. Kelly
- Division of Biomedical EngineeringUniversity of Saskatchewan57 Campus DriveSaskatoonS7N 5A9Canada
- Department of Surgery, College of MedicineUniversity of Saskatchewan107 Wiggins RoadSaskatoonS7N 5E5Canada
| | - Ildiko Badea
- College of Pharmacy and NutritionUniversity of Saskatchewan107 Wiggins RoadSaskatoonS7N 5E5SaskatchewanCanada
| | - Xiongbiao Chen
- Division of Biomedical EngineeringUniversity of Saskatchewan57 Campus DriveSaskatoonS7N 5A9Canada
- Department of Mechanical EngineeringUniversity of Saskatchewan57 Campus DriveSaskatoonS7N 5A9Canada
| |
Collapse
|
10
|
Lin S, Yang Y, Zhou Z, Li W, Wang X, Liu Y, Bi Y, Mao J. Regulation mechanism of microRNAs in cardiac cells-derived exosomes in cell crosstalk. Front Pharmacol 2024; 15:1399850. [PMID: 39228519 PMCID: PMC11368792 DOI: 10.3389/fphar.2024.1399850] [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: 03/12/2024] [Accepted: 07/22/2024] [Indexed: 09/05/2024] Open
Abstract
The heart is a multicellular system, and the intercellular crosstalk mechanism is very important for the growth and development of the heart and even the organs, tissues, and cells at a distance. As a kind of extracellular vesicle, exosomes are released by different types of cells and can carry specific genetic material, endosomal proteins, cytokines, etc., which are the main material basis for mediating cell crosstalk mechanism. Among them, microRNA carried by cardiac cells-derived exosomes have highly conserved sequences and play a key role in regulating the function of organs, tissues, and cells related to cardiovascular diseases and their complications and comorbidities, which have attracted extensive attention in the medical community in recent years. Following up on the latest research progress at home and abroad, this review systematically summarized the regulatory role of microRNA from cardiac cells-derived exosomes in various cell crosstalk, including not only cardiac cells (including cardiomyocytes, fibroblasts, myofibroblast, cardiac progenitor cells, cardiac microvascular endothelial cells, cardiosphere-derived cells, etc.) but also tumor cells, bone marrow progenitor cells, and other tissue cells, in order to provide a reference for the prevention and treatment of cardiovascular diseases and their complications and comorbidities.
Collapse
Affiliation(s)
- Shanshan Lin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuanjian Yang
- Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Zhou Zhou
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wen Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yu Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yingfei Bi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
11
|
Ege D, Lu HH, Boccaccini AR. Bioactive Glass and Silica Particles for Skeletal and Cardiac Muscle Tissue Regeneration. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:448-461. [PMID: 38126329 DOI: 10.1089/ten.teb.2023.0277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
When skeletal and cardiac tissues are damaged, surgical approaches are not always successful and tissue regeneration approaches are investigated. Reports in the literature indicate that silica nanoparticles and bioactive glasses (BGs), including silicate bioactive glasses (e.g., 45S5 BG), phosphate glass fibers, boron-doped mesoporous BGs, borosilicate glasses, and aluminoborates, are promising for repairing skeletal muscle tissue. Silica nanoparticles and BGs have been combined with polymers to obtain aligned nanofibers and to maintain controlled delivery of nanoparticles for skeletal muscle repair. The literature indicates that cardiac muscle regeneration can be also triggered by the ionic products of BGs. This was observed to be due to the release of vascular endothelial growth factor and other growth factors from cardiomyocytes, which regulate endothelial cells to form capillary structures (angiogenesis). Specific studies, including both in vitro and in vivo approaches, are reviewed in this article. The analysis of the literature indicates that although the research field is still very limited, BGs are showing great promise for muscle tissue engineering and further research in the field should be carried out to expand our basic knowledge on the application of BGs in muscle (skeletal and cardiac) tissue regeneration. Impact statement This review highlights the potential of silica particles and bioactive glasses (BGs) for skeletal and cardiac tissue regeneration. These biomaterials create scaffolds triggering muscle cell differentiation. Ionic products from BGs stimulate growth factors, supporting angiogenesis in cardiac tissue repair. Further research is required to expand our know-how on silica particles and BGs in muscle tissue engineering.
Collapse
Affiliation(s)
- Duygu Ege
- Institute of Biomedical Engineering, Bogazici University, Istanbul, Turkey
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Hsuan-Heng Lu
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Aldo R Boccaccini
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
12
|
Cofiño-Fabres C, Boonen T, Rivera-Arbeláez JM, Rijpkema M, Blauw L, Rensen PCN, Schwach V, Ribeiro MC, Passier R. Micro-Engineered Heart Tissues On-Chip with Heterotypic Cell Composition Display Self-Organization and Improved Cardiac Function. Adv Healthc Mater 2024; 13:e2303664. [PMID: 38471185 DOI: 10.1002/adhm.202303664] [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: 10/26/2023] [Revised: 01/30/2024] [Indexed: 03/14/2024]
Abstract
Advanced in vitro models that recapitulate the structural organization and function of the human heart are highly needed for accurate disease modeling, more predictable drug screening, and safety pharmacology. Conventional 3D Engineered Heart Tissues (EHTs) lack heterotypic cell complexity and culture under flow, whereas microfluidic Heart-on-Chip (HoC) models in general lack the 3D configuration and accurate contractile readouts. In this study, an innovative and user-friendly HoC model is developed to overcome these limitations, by culturing human pluripotent stem cell (hPSC)-derived cardiomyocytes (CMs), endothelial (ECs)- and smooth muscle cells (SMCs), together with human cardiac fibroblasts (FBs), underflow, leading to self-organized miniaturized micro-EHTs (µEHTs) with a CM-EC interface reminiscent of the physiological capillary lining. µEHTs cultured under flow display enhanced contractile performance and conduction velocity. In addition, the presence of the EC layer altered drug responses in µEHT contraction. This observation suggests a potential barrier-like function of ECs, which may affect the availability of drugs to the CMs. These cardiac models with increased physiological complexity, will pave the way to screen for therapeutic targets and predict drug efficacy.
Collapse
Affiliation(s)
- Carla Cofiño-Fabres
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, 7522 NB, The Netherlands
| | - Tom Boonen
- River BioMedics B.V, Enschede, 7522 NB, The Netherlands
| | - José M Rivera-Arbeláez
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, 7522 NB, The Netherlands
- BIOS Lab-on-a-Chip Group, MESA+ Institute for Nanotechnology, Max Planck Institute for Complex Fluid Dynamics, University of Twente, Enschede, 7522 NB, The Netherlands
| | - Minke Rijpkema
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, 2300 RC, The Netherlands
| | - Lisanne Blauw
- River BioMedics B.V, Enschede, 7522 NB, The Netherlands
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, 2300 RC, The Netherlands
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, 2300 RC, The Netherlands
| | - Verena Schwach
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, 7522 NB, The Netherlands
| | - Marcelo C Ribeiro
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, 7522 NB, The Netherlands
- River BioMedics B.V, Enschede, 7522 NB, The Netherlands
| | - Robert Passier
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, 7522 NB, The Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, 2300 RC, The Netherlands
| |
Collapse
|
13
|
Chen T, Ellman DG, Fang S, Bak ST, Nørgård MØ, Svenningsen P, Andersen DC. Transfer of cardiomyocyte-derived extracellular vesicles to neighboring cardiac cells requires tunneling nanotubes during heart development. Theranostics 2024; 14:3843-3858. [PMID: 38994028 PMCID: PMC11234280 DOI: 10.7150/thno.91604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 05/26/2024] [Indexed: 07/13/2024] Open
Abstract
Rationale: Extracellular vesicles (EVs) are thought to mediate intercellular communication during development and disease. Yet, biological insight to intercellular EV transfer remains elusive, also in the heart, and is technically challenging to demonstrate. Here, we aimed to investigate biological transfer of cardiomyocyte-derived EVs in the neonatal heart. Methods: We exploited CD9 as a marker of EVs, and generated two lines of cardiomyocyte specific EV reporter mice: Tnnt2-Cre; double-floxed inverted CD9/EGFP and αMHC-MerCreMer; double-floxed inverted CD9/EGFP. The two mouse lines were utilized to determine whether developing cardiomyocytes transfer EVs to other cardiac cells (non-myocytes and cardiomyocytes) in vitro and in vivo and investigate the intercellular transport pathway of cardiomyocyte-derived EVs. Results: Genetic tagging of cardiomyocytes was confirmed in both reporter mouse lines and proof of concept in the postnatal heart showed that, a fraction of EGFP+/MYH1- non-myocytes exist firmly demonstrating in vivo cardiomyocyte-derived EV transfer. However, two sets of direct and indirect EGFP +/- cardiac cell co-cultures showed that cardiomyocyte-derived EGFP+ EV transfer requires cell-cell contact and that uptake of EGFP+ EVs from the medium is limited. The same was observed when co-cultiring with mouse macrophages. Further mechanistic insight showed that cardiomyocyte EV transfer occurs through type I tunneling nanotubes. Conclusion: While the current notion assumes that EVs are transferred through secretion to the surroundings, our data show that cardiomyocyte-derived EV transfer in the developing heart occurs through nanotubes between neighboring cells. Whether these data are fundamental and relate to adult hearts and other organs remains to be determined, but they imply that the normal developmental process of EV transfer goes through cell-cell contact rather than through the extracellular compartment.
Collapse
Affiliation(s)
- Ting Chen
- Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Urology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Ditte Gry Ellman
- Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Shu Fang
- Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Sara Thornby Bak
- Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mikkel Ørnfeldt Nørgård
- Department of Molecular Medicine, Unit of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Per Svenningsen
- Department of Molecular Medicine, Unit of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Ditte Caroline Andersen
- Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
14
|
Andrée B, Voß N, Kriedemann N, Triebert W, Teske J, Mertens M, Witte M, Szádocka S, Hilfiker A, Aper T, Gruh I, Zweigerdt R. Fabrication of heart tubes from iPSC derived cardiomyocytes and human fibrinogen by rotating mold technology. Sci Rep 2024; 14:13174. [PMID: 38849457 PMCID: PMC11161509 DOI: 10.1038/s41598-024-64022-7] [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: 03/06/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024] Open
Abstract
Due to its structural and functional complexity the heart imposes immense physical, physiological and electromechanical challenges on the engineering of a biological replacement. Therefore, to come closer to clinical translation, the development of a simpler biological assist device is requested. Here, we demonstrate the fabrication of tubular cardiac constructs with substantial dimensions of 6 cm in length and 11 mm in diameter by combining human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) and human foreskin fibroblast (hFFs) in human fibrin employing a rotating mold technology. By centrifugal forces employed in the process a cell-dense layer was generated enabling a timely functional coupling of iPSC-CMs demonstrated by a transgenic calcium sensor, rhythmic tissue contractions, and responsiveness to electrical pacing. Adjusting the degree of remodeling as a function of hFF-content and inhibition of fibrinolysis resulted in stable tissue integrity for up to 5 weeks. The rotating mold device developed in frame of this work enabled the production of tubes with clinically relevant dimensions of up to 10 cm in length and 22 mm in diameter which-in combination with advanced bioreactor technology for controlled production of functional iPSC-derivatives-paves the way towards the clinical translation of a biological cardiac assist device.
Collapse
Affiliation(s)
- Birgit Andrée
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, MHH-Hannover Medical School, Carl Neuberg Str. 1, 30625, Hannover, Germany.
| | - Nils Voß
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, MHH-Hannover Medical School, Carl Neuberg Str. 1, 30625, Hannover, Germany
| | - Nils Kriedemann
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, MHH-Hannover Medical School, Carl Neuberg Str. 1, 30625, Hannover, Germany
| | - Wiebke Triebert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, MHH-Hannover Medical School, Carl Neuberg Str. 1, 30625, Hannover, Germany
| | - Jana Teske
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, MHH-Hannover Medical School, Carl Neuberg Str. 1, 30625, Hannover, Germany
| | - Mira Mertens
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, MHH-Hannover Medical School, Carl Neuberg Str. 1, 30625, Hannover, Germany
| | - Merlin Witte
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, MHH-Hannover Medical School, Carl Neuberg Str. 1, 30625, Hannover, Germany
| | - Sára Szádocka
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, MHH-Hannover Medical School, Carl Neuberg Str. 1, 30625, Hannover, Germany
| | - Andres Hilfiker
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, MHH-Hannover Medical School, Carl Neuberg Str. 1, 30625, Hannover, Germany
| | - Thomas Aper
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, MHH-Hannover Medical School, Carl Neuberg Str. 1, 30625, Hannover, Germany
| | - Ina Gruh
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, MHH-Hannover Medical School, Carl Neuberg Str. 1, 30625, Hannover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, MHH-Hannover Medical School, Carl Neuberg Str. 1, 30625, Hannover, Germany
| |
Collapse
|
15
|
Cadosch N, Gil-Cruz C, Perez-Shibayama C, Ludewig B. Cardiac Fibroblastic Niches in Homeostasis and Inflammation. Circ Res 2024; 134:1703-1717. [PMID: 38843287 PMCID: PMC11149942 DOI: 10.1161/circresaha.124.323892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 06/09/2024]
Abstract
Fibroblasts are essential for building and maintaining the structural integrity of all organs. Moreover, fibroblasts can acquire an inflammatory phenotype to accommodate immune cells in specific niches and to provide migration, differentiation, and growth factors. In the heart, balancing of fibroblast activity is critical for cardiac homeostasis and optimal organ function during inflammation. Fibroblasts sustain cardiac homeostasis by generating local niche environments that support housekeeping functions and by actively engaging in intercellular cross talk. During inflammatory perturbations, cardiac fibroblasts rapidly switch to an inflammatory state and actively communicate with infiltrating immune cells to orchestrate immune cell migration and activity. Here, we summarize the current knowledge on the molecular landscape of cardiac fibroblasts, focusing on their dual role in promoting tissue homeostasis and modulating immune cell-cardiomyocyte interaction. In addition, we discuss potential future avenues for manipulating cardiac fibroblast activity during myocardial inflammation.
Collapse
Affiliation(s)
- Nadine Cadosch
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland (N.C., C.G.-C., C.P.-S., B.L.)
| | - Cristina Gil-Cruz
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland (N.C., C.G.-C., C.P.-S., B.L.)
- University Heart Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland (C.G.-C., B.L.), University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Christian Perez-Shibayama
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland (N.C., C.G.-C., C.P.-S., B.L.)
| | - Burkhard Ludewig
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland (N.C., C.G.-C., C.P.-S., B.L.)
- University Heart Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland (C.G.-C., B.L.), University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Center for Translational and Experimental Cardiology (B.L.), University Hospital Zurich and University of Zurich, Zurich, Switzerland
| |
Collapse
|
16
|
Climent M, García-Giménez JL. Special Issue "The Role of Non-Coding RNAs Involved in Cardiovascular Diseases and Cellular Communication". Int J Mol Sci 2024; 25:6034. [PMID: 38892220 PMCID: PMC11172417 DOI: 10.3390/ijms25116034] [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: 04/16/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Despite the great progress in diagnosis, prevention, and treatment, cardiovascular diseases (CVDs) are still the most prominent cause of death worldwide [...].
Collapse
Affiliation(s)
- Montserrat Climent
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - José Luis García-Giménez
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain;
- Health Research Institute INCLIVA, 46010 Valencia, Spain
- Center for Biomedical Research Network on Rare Diseases (CIBERER), Carlos III Health Institute, 46010 Valencia, Spain
| |
Collapse
|
17
|
Lu Y, Chen QM, An L. SPADE: spatial deconvolution for domain specific cell-type estimation. Commun Biol 2024; 7:469. [PMID: 38632414 PMCID: PMC11024133 DOI: 10.1038/s42003-024-06172-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] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
Understanding gene expression in different cell types within their spatial context is a key goal in genomics research. SPADE (SPAtial DEconvolution), our proposed method, addresses this by integrating spatial patterns into the analysis of cell type composition. This approach uses a combination of single-cell RNA sequencing, spatial transcriptomics, and histological data to accurately estimate the proportions of cell types in various locations. Our analyses of synthetic data have demonstrated SPADE's capability to discern cell type-specific spatial patterns effectively. When applied to real-life datasets, SPADE provides insights into cellular dynamics and the composition of tumor tissues. This enhances our comprehension of complex biological systems and aids in exploring cellular diversity. SPADE represents a significant advancement in deciphering spatial gene expression patterns, offering a powerful tool for the detailed investigation of cell types in spatial transcriptomics.
Collapse
Affiliation(s)
- Yingying Lu
- Interdisciplinary Program in Statistics and Data Science, University of Arizona, Tucson, AZ, 85721, USA
| | - Qin M Chen
- College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Lingling An
- Interdisciplinary Program in Statistics and Data Science, University of Arizona, Tucson, AZ, 85721, USA.
- Department of Biosystems Engineering, University of Arizona, Tucson, AZ, 85721, USA.
- Department of Epidemiology and Biostatistics, University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
18
|
Singh S, Paul D, Nath V, A R. Exosomes: current knowledge and future perspectives. Tissue Barriers 2024; 12:2232248. [PMID: 37439246 PMCID: PMC11042064 DOI: 10.1080/21688370.2023.2232248] [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: 05/08/2023] [Accepted: 06/27/2023] [Indexed: 07/14/2023] Open
Abstract
Exosomes are membrane-bound micro-vesicles that possess endless therapeutic potential for treatment of numerous pathologies including autoimmune, cardiovascular, ocular, and nervous disorders. Despite considerable knowledge about exosome biogenesis and secretion, still, there is a lack of information regarding exosome uptake by cell types and internal signaling pathways through which these exosomes process cellular response. Exosomes are key components of cell signaling and intercellular communication. In central nervous system (CNS), exosomes can penetrate BBB and maintain homeostasis by myelin sheath regulation and the waste products elimination. Therefore, the current review summarizes role of exosomes and their use as biomarkers in cardiovascular, nervous and ocular disorders. This aspect of exosomes provides positive hope to monitor disease development and enable early diagnosis and treatment optimization. In this review, we have summarized recent findings on physiological and therapeutic effects of exosomes and also attempt to provide insights about stress-preconditioned exosomes and stem cell-derived exosomes.
Collapse
Affiliation(s)
- Swati Singh
- College of Pharmacy, JSS Academy of Technical Sciences, Noida, Uttar Pradesh, India
| | - Deepraj Paul
- College of Pharmacy, JSS Academy of Technical Sciences, Noida, Uttar Pradesh, India
| | - Virendra Nath
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Ajmer, India
| | - Rohini A
- College of Pharmacy, JSS Academy of Technical Sciences, Noida, Uttar Pradesh, India
| |
Collapse
|
19
|
Liu X, Li B, Wang S, Zhang E, Schultz M, Touma M, Monteiro Da Rocha A, Evans SM, Eichmann A, Herron T, Chen R, Xiong D, Jaworski A, Weiss S, Si MS. Stromal Cell-SLIT3/Cardiomyocyte-ROBO1 Axis Regulates Pressure Overload-Induced Cardiac Hypertrophy. Circ Res 2024; 134:913-930. [PMID: 38414132 PMCID: PMC10977056 DOI: 10.1161/circresaha.122.321292] [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: 04/27/2022] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND Recently shown to regulate cardiac development, the secreted axon guidance molecule SLIT3 maintains its expression in the postnatal heart. Despite its known expression in the cardiovascular system after birth, SLIT3's relevance to cardiovascular function in the postnatal state remains unknown. As such, the objectives of this study were to determine the postnatal myocardial sources of SLIT3 and to evaluate its functional role in regulating the cardiac response to pressure overload stress. METHODS We performed in vitro studies on cardiomyocytes and myocardial tissue samples from patients and performed in vivo investigation with SLIT3 and ROBO1 (roundabout homolog 1) mutant mice undergoing transverse aortic constriction to establish the role of SLIT3-ROBO1 in adverse cardiac remodeling. RESULTS We first found that SLIT3 transcription was increased in myocardial tissue obtained from patients with congenital heart defects that caused ventricular pressure overload. Immunostaining of hearts from WT (wild-type) and reporter mice revealed that SLIT3 is secreted by cardiac stromal cells, namely fibroblasts and vascular mural cells, within the heart. Conditioned media from cardiac fibroblasts and vascular mural cells both stimulated cardiomyocyte hypertrophy in vitro, an effect that was partially inhibited by an anti-SLIT3 antibody. Also, the N-terminal, but not the C-terminal, fragment of SLIT3 and the forced overexpression of SLIT3 stimulated cardiomyocyte hypertrophy and the transcription of hypertrophy-related genes. We next determined that ROBO1 was the most highly expressed roundabout receptor in cardiomyocytes and that ROBO1 mediated SLIT3's hypertrophic effects in vitro. In vivo, Tcf21+ fibroblast and Tbx18+ vascular mural cell-specific knockout of SLIT3 in mice resulted in decreased left ventricular hypertrophy and cardiac fibrosis after transverse aortic constriction. Furthermore, α-MHC+ cardiomyocyte-specific deletion of ROBO1 also preserved left ventricular function and abrogated hypertrophy, but not fibrosis, after transverse aortic constriction. CONCLUSIONS Collectively, these results indicate a novel role for the SLIT3-ROBO1-signaling axis in regulating postnatal cardiomyocyte hypertrophy induced by pressure overload.
Collapse
Affiliation(s)
- Xiaoxiao Liu
- Department of Cardiac Surgery (X.L., B.L., S.W., D.X., M.-S.S.), Michigan Medicine, Ann Arbor
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, China (X.L., R.C.)
| | - Baolei Li
- Department of Cardiac Surgery (X.L., B.L., S.W., D.X., M.-S.S.), Michigan Medicine, Ann Arbor
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, China (B.L.)
| | - Shuyun Wang
- Department of Cardiac Surgery (X.L., B.L., S.W., D.X., M.-S.S.), Michigan Medicine, Ann Arbor
| | - Erge Zhang
- Division of Cardiac Surgery, Department of Surgery (E.Z., M.S., M.-S.S.), David Geffen School of Medicine University of California, Los Angeles
| | - Megan Schultz
- Division of Cardiac Surgery, Department of Surgery (E.Z., M.S., M.-S.S.), David Geffen School of Medicine University of California, Los Angeles
| | - Marlin Touma
- Department of Pediatrics (M.T.), David Geffen School of Medicine University of California, Los Angeles
| | - Andre Monteiro Da Rocha
- Division of Cardiovascular Medicine, Department of Internal Medicine (A.M.D.R., T.H.), Michigan Medicine, Ann Arbor
| | - Sylvia M. Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences (S.M.E.), University of California, San Diego, La Jolla
- Department of Medicine, School of Medicine (S.M.E.), University of California, San Diego, La Jolla
| | - Anne Eichmann
- Department of Internal Medicine, Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.E.)
- INSERM, Paris Cardiovascular Research Center (PARCC), Université de Paris, France (A.E.)
| | - Todd Herron
- Division of Cardiovascular Medicine, Department of Internal Medicine (A.M.D.R., T.H.), Michigan Medicine, Ann Arbor
| | - Ruizhen Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, China (X.L., R.C.)
| | - Dingding Xiong
- Department of Cardiac Surgery (X.L., B.L., S.W., D.X., M.-S.S.), Michigan Medicine, Ann Arbor
| | - Alexander Jaworski
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI (A.J.)
| | - Stephen Weiss
- Life Sciences Institute, University of Michigan, Ann Arbor (S.W.)
| | - Ming-Sing Si
- Department of Cardiac Surgery (X.L., B.L., S.W., D.X., M.-S.S.), Michigan Medicine, Ann Arbor
- Division of Cardiac Surgery, Department of Surgery (E.Z., M.S., M.-S.S.), David Geffen School of Medicine University of California, Los Angeles
| |
Collapse
|
20
|
Chi C, Roland TJ, Song K. Differentiation of Pluripotent Stem Cells for Disease Modeling: Learning from Heart Development. Pharmaceuticals (Basel) 2024; 17:337. [PMID: 38543122 PMCID: PMC10975450 DOI: 10.3390/ph17030337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 04/01/2024] Open
Abstract
Heart disease is a pressing public health problem and the leading cause of death worldwide. The heart is the first organ to gain function during embryogenesis in mammals. Heart development involves cell determination, expansion, migration, and crosstalk, which are orchestrated by numerous signaling pathways, such as the Wnt, TGF-β, IGF, and Retinoic acid signaling pathways. Human-induced pluripotent stem cell-based platforms are emerging as promising approaches for modeling heart disease in vitro. Understanding the signaling pathways that are essential for cardiac development has shed light on the molecular mechanisms of congenital heart defects and postnatal heart diseases, significantly advancing stem cell-based platforms to model heart diseases. This review summarizes signaling pathways that are crucial for heart development and discusses how these findings improve the strategies for modeling human heart disease in vitro.
Collapse
Affiliation(s)
- Congwu Chi
- Heart Institute, University of South Florida, Tampa, FL 33602, USA; (C.C.); (T.J.R.)
- Department of Internal Medicine, University of South Florida, Tampa, FL 33602, USA
- Center for Regenerative Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Truman J. Roland
- Heart Institute, University of South Florida, Tampa, FL 33602, USA; (C.C.); (T.J.R.)
- Department of Internal Medicine, University of South Florida, Tampa, FL 33602, USA
- Center for Regenerative Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Kunhua Song
- Heart Institute, University of South Florida, Tampa, FL 33602, USA; (C.C.); (T.J.R.)
- Department of Internal Medicine, University of South Florida, Tampa, FL 33602, USA
- Center for Regenerative Medicine, University of South Florida, Tampa, FL 33602, USA
| |
Collapse
|
21
|
Butler D, Reyes DR. Heart-on-a-chip systems: disease modeling and drug screening applications. LAB ON A CHIP 2024; 24:1494-1528. [PMID: 38318723 DOI: 10.1039/d3lc00829k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide, casting a substantial economic footprint and burdening the global healthcare system. Historically, pre-clinical CVD modeling and therapeutic screening have been performed using animal models. Unfortunately, animal models oftentimes fail to adequately mimic human physiology, leading to a poor translation of therapeutics from pre-clinical trials to consumers. Even those that make it to market can be removed due to unforeseen side effects. As such, there exists a clinical, technological, and economical need for systems that faithfully capture human (patho)physiology for modeling CVD, assessing cardiotoxicity, and evaluating drug efficacy. Heart-on-a-chip (HoC) systems are a part of the broader organ-on-a-chip paradigm that leverages microfluidics, tissue engineering, microfabrication, electronics, and gene editing to create human-relevant models for studying disease, drug-induced side effects, and therapeutic efficacy. These compact systems can be capable of real-time measurements and on-demand characterization of tissue behavior and could revolutionize the drug development process. In this review, we highlight the key components that comprise a HoC system followed by a review of contemporary reports of their use in disease modeling, drug toxicity and efficacy assessment, and as part of multi-organ-on-a-chip platforms. We also discuss future perspectives and challenges facing the field, including a discussion on the role that standardization is expected to play in accelerating the widespread adoption of these platforms.
Collapse
Affiliation(s)
- Derrick Butler
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA.
| | - Darwin R Reyes
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA.
| |
Collapse
|
22
|
Kiessling P, Kuppe C. Spatial multi-omics: novel tools to study the complexity of cardiovascular diseases. Genome Med 2024; 16:14. [PMID: 38238823 PMCID: PMC10795303 DOI: 10.1186/s13073-024-01282-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 01/02/2024] [Indexed: 01/22/2024] Open
Abstract
Spatial multi-omic studies have emerged as a promising approach to comprehensively analyze cells in tissues, enabling the joint analysis of multiple data modalities like transcriptome, epigenome, proteome, and metabolome in parallel or even the same tissue section. This review focuses on the recent advancements in spatial multi-omics technologies, including novel data modalities and computational approaches. We discuss the advancements in low-resolution and high-resolution spatial multi-omics methods which can resolve up to 10,000 of individual molecules at subcellular level. By applying and integrating these techniques, researchers have recently gained valuable insights into the molecular circuits and mechanisms which govern cell biology along the cardiovascular disease spectrum. We provide an overview of current data analysis approaches, with a focus on data integration of multi-omic datasets, highlighting strengths and weaknesses of various computational pipelines. These tools play a crucial role in analyzing and interpreting spatial multi-omics datasets, facilitating the discovery of new findings, and enhancing translational cardiovascular research. Despite nontrivial challenges, such as the need for standardization of experimental setups, data analysis, and improved computational tools, the application of spatial multi-omics holds tremendous potential in revolutionizing our understanding of human disease processes and the identification of novel biomarkers and therapeutic targets. Exciting opportunities lie ahead for the spatial multi-omics field and will likely contribute to the advancement of personalized medicine for cardiovascular diseases.
Collapse
Affiliation(s)
- Paul Kiessling
- Department of Nephrology, Rheumatology, and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
| | - Christoph Kuppe
- Department of Nephrology, Rheumatology, and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany.
| |
Collapse
|
23
|
Gao J, Mo S, Wang J, Zhang M, Shi Y, Zhu C, Shang Y, Tang X, Zhang S, Wu X, Xu X, Wang Y, Li Z, Zheng G, Chen Z, Wang Q, Tang K, Cao Z. MACC: a visual interactive knowledgebase of metabolite-associated cell communications. Nucleic Acids Res 2024; 52:D633-D639. [PMID: 37897362 PMCID: PMC10767829 DOI: 10.1093/nar/gkad914] [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: 08/15/2023] [Revised: 09/21/2023] [Accepted: 10/10/2023] [Indexed: 10/30/2023] Open
Abstract
Metabolite-associated cell communications play critical roles in maintaining the normal biological function of human through coordinating cells, organs and physiological systems. Though substantial information of MACCs has been continuously reported, no relevant database has become available so far. To address this gap, we here developed the first knowledgebase (MACC), to comprehensively describe human metabolite-associated cell communications through curation of experimental literatures. MACC currently contains: (a) 4206 carefully curated metabolite-associated cell communications pairs involving 244 human endogenous metabolites and reported biological effects in vivo and in vitro; (b) 226 comprehensive cell subtypes and 296 disease states, such as cancers, autoimmune diseases, and pathogenic infections; (c) 4508 metabolite-related enzymes and transporters, involving 542 pathways; (d) an interactive tool with user-friendly interface to visualize networks of multiple metabolite-cell interactions. (e) overall expression landscape of metabolite-associated gene sets derived from over 1500 single-cell expression profiles to infer metabolites variations across different cells in the sample. Also, MACC enables cross-links to well-known databases, such as HMDB, DrugBank, TTD and PubMed etc. In complement to ligand-receptor databases, MACC may give new perspectives of alternative communication between cells via metabolite secretion and adsorption, together with the resulting biological functions. MACC is publicly accessible at: http://macc.badd-cao.net/.
Collapse
Affiliation(s)
- Jian Gao
- School of Life Sciences, Fudan University, Shanghai, China
- International Human Phenome Institutes (Shanghai), Shanghai, China
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Saifeng Mo
- Dept. of Gastroenterology, Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jun Wang
- School of Life Sciences, Fudan University, Shanghai, China
| | - Mou Zhang
- School of Life Sciences, Fudan University, Shanghai, China
| | - Yao Shi
- School of Life Sciences, Fudan University, Shanghai, China
| | - Chuhan Zhu
- Dept. of Gastroenterology, Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yuxuan Shang
- Biological Sciences, University of California Santa Barbara, CA, USA
| | - Xinyue Tang
- Dept. of Gastroenterology, Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Shiyue Zhang
- School of Life Sciences, Fudan University, Shanghai, China
| | - Xinwen Wu
- Dept. of Gastroenterology, Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xinyan Xu
- Dept. of Gastroenterology, Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yiheng Wang
- School of Life Sciences, Fudan University, Shanghai, China
| | - Zihao Li
- Dept. of Gastroenterology, Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Genhui Zheng
- Dept. of Gastroenterology, Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zikun Chen
- Dept. of Gastroenterology, Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Qiming Wang
- School of Life Sciences, Fudan University, Shanghai, China
| | - Kailin Tang
- Dept. of Gastroenterology, Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zhiwei Cao
- School of Life Sciences, Fudan University, Shanghai, China
- International Human Phenome Institutes (Shanghai), Shanghai, China
| |
Collapse
|
24
|
Dobreva G, Heineke J. Inter- and Intracellular Signaling Pathways. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:271-294. [PMID: 38884717 DOI: 10.1007/978-3-031-44087-8_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Cardiovascular diseases, both congenital and acquired, are the leading cause of death worldwide, associated with significant health consequences and economic burden. Due to major advances in surgical procedures, most patients with congenital heart disease (CHD) survive into adulthood but suffer from previously unrecognized long-term consequences, such as early-onset heart failure. Therefore, understanding the molecular mechanisms resulting in heart defects and the lifelong complications due to hemodynamic overload are of utmost importance. Congenital heart disease arises in the first trimester of pregnancy, due to defects in the complex morphogenetic patterning of the heart. This process is coordinated through a complicated web of intercellular communication between the epicardium, the endocardium, and the myocardium. In the postnatal heart, similar crosstalk between cardiomyocytes, endothelial cells, and fibroblasts exists during pathological hemodynamic overload that emerges as a consequence of a congenital heart defect. Ultimately, communication between cells triggers the activation of intracellular signaling circuits, which allow fine coordination of cardiac development and function. Here, we review the inter- and intracellular signaling mechanisms in the heart as they were discovered mainly in genetically modified mice.
Collapse
Affiliation(s)
- Gergana Dobreva
- ECAS (European Center for Angioscience), Department of Cardiovascular Genomics and Epigenomics, Mannheim Faculty of Medicine, Heidelberg University, Mannheim, Germany.
- German Centre for Cardiovascular Research (DZHK) Partner Site, Heidelberg/Mannheim, Germany.
| | - Joerg Heineke
- German Centre for Cardiovascular Research (DZHK) Partner Site, Heidelberg/Mannheim, Germany.
- ECAS (European Center for Angioscience), Department of Cardiovascular Physiology, Mannheim Faculty of Medicine, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
25
|
Orgil BO, Purevjav E. Molecular Pathways and Animal Models of Cardiomyopathies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:991-1019. [PMID: 38884766 DOI: 10.1007/978-3-031-44087-8_64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Cardiomyopathies are a heterogeneous group of disorders of the heart muscle that ultimately result in congestive heart failure. Rapid progress in genetics, molecular and cellular biology with breakthrough innovative genetic-engineering techniques, such as next-generation sequencing and multiomics platforms, stem cell reprogramming, as well as novel groundbreaking gene-editing systems over the past 25 years has greatly improved the understanding of pathogenic signaling pathways in inherited cardiomyopathies. This chapter will focus on intracellular and intercellular molecular signaling pathways that are activated by a genetic insult in cardiomyocytes to maintain tissue and organ level regulation and resultant cardiac remodeling in certain forms of cardiomyopathies. In addition, animal models of different clinical forms of human cardiomyopathies with their summaries of triggered key molecules and signaling pathways will be described.
Collapse
Affiliation(s)
- Buyan-Ochir Orgil
- Department of Pediatrics, The Heart Institute, Division of Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Enkhsaikhan Purevjav
- Department of Pediatrics, The Heart Institute, Division of Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
26
|
Pradeep SR, Thirunavukkarasu M, Accorsi D, Swaminathan S, Lim ST, Cernuda B, Kemerley A, Hubbard J, Campbell J, Wilson RL, Coca-Soliz V, Tapias L, Selvaraju V, Jellison ER, Yee SP, Palesty JA, Maulik N. Novel approaches to determine the functional role of cardiomyocyte specific E3 ligase, Pellino-1 following myocardial infarction. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166899. [PMID: 37778482 DOI: 10.1016/j.bbadis.2023.166899] [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: 05/26/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
OBJECTIVES Ubiquitination plays a vital role in controlling vascular inflammation, cellular protein quality control, and minimizing misfolded protein toxicity. Pellino-1 (Peli1), a type of E3 ubiquitin ligase, has emerged as a critical regulator of the innate immune response; however, its role in the repair and regeneration of ischemic myocardium remains to be elucidated. METHODS Mice (8-12 weeks old, male and females) were divided into (i) Wild type (ii) cardiomyocyte-specific Peli1 overexpressed (AMPEL1Tg/+), (iii) cardiomyocyte-specific Peli1 knockout (CP1KO) and were subjected to sham and left anterior descending artery ligation. The tissues were collected at various time points after surgery for Western blot, and immunohistochemical analyses. Echocardiography is performed 30 days after myocardial infarction. Cardiomyocytes isolated from wild-type, Peli1 overexpressed and knockout mice were used to study the interaction between cardiomyocytes and endothelial cells in vitro under oxidative stress and cells were used for Western blot, flow cytometric analysis, and scratch assay. RESULTS We observed faster wound closure and increased expression of angiogenic factors with MCECs treated with conditioned media obtained from the AMPEL1Tg/+ cardiomyocytes compared to CPIKO and WT cardiomyocytes. Again, AMPEL1Tg/+MI mice showed preserved systolic function and reduced fibrosis compared to the CPIKOMI and WTMI groups. Capillary and arteriolar density were found to be increased in AMPEL1Tg/+MI compared to CP1KOMI. Increased survival and angiogenic factors such as p-Akt, p-MK2, p-IkBα, VEGF, cIAP2, and Bcl2 were observed in AMPEL1Tg/+ compared to CP1KO and WT mice subjected to MI. CONCLUSION The present study uncovers the crucial role of cardiac Peli1 as a regulator of the repair and regeneration of ischemic myocardium by using multiple genetically engineered mouse models.
Collapse
Affiliation(s)
- Seetur R Pradeep
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA
| | - Mahesh Thirunavukkarasu
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA
| | - Diego Accorsi
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Santosh Swaminathan
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Sue Ting Lim
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Bryan Cernuda
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA
| | - Andrew Kemerley
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA
| | - Jennifer Hubbard
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Jacob Campbell
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA
| | - Rickesha L Wilson
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA
| | - Vladimir Coca-Soliz
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Leonidas Tapias
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA; Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Vaithinathan Selvaraju
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA
| | - Evan R Jellison
- Department of Immunology, University of Connecticut Health, School of Medicine, Farmington, CT, USA
| | - Siu-Pok Yee
- Center for Mouse Genome Modification, University of Connecticut Health, School of Medicine, Farmington, CT, USA
| | - J Alexander Palesty
- Stanley J. Dudrick, Department of Surgery, Saint Mary's Hospital, Waterbury 06706, CT, USA
| | - Nilanjana Maulik
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, School of Medicine, Farmington 06030, CT, USA.
| |
Collapse
|
27
|
Lin CY, Chang YM, Tseng HY, Shih YL, Yeh HH, Liao YR, Tang HH, Hsu CL, Chen CC, Yan YT, Kao CF. Epigenetic regulator RNF20 underlies temporal hierarchy of gene expression to regulate postnatal cardiomyocyte polarization. Cell Rep 2023; 42:113416. [PMID: 37967007 DOI: 10.1016/j.celrep.2023.113416] [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: 12/08/2022] [Revised: 09/19/2023] [Accepted: 10/25/2023] [Indexed: 11/17/2023] Open
Abstract
Differentiated cardiomyocytes (CMs) must undergo diverse morphological and functional changes during postnatal development. However, the mechanisms underlying initiation and coordination of these changes remain unclear. Here, we delineate an integrated, time-ordered transcriptional network that begins with expression of genes for cell-cell connections and leads to a sequence of structural, cell-cycle, functional, and metabolic transitions in mouse postnatal hearts. Depletion of histone H2B ubiquitin ligase RNF20 disrupts this gene network and impairs CM polarization. Subsequently, assay for transposase-accessible chromatin using sequencing (ATAC-seq) analysis confirmed that RNF20 contributes to chromatin accessibility in this context. As such, RNF20 is likely to facilitate binding of transcription factors at the promoters of genes involved in cell-cell connections and actin organization, which are crucial for CM polarization and functional integration. These results suggest that CM polarization is one of the earliest events during postnatal heart development and provide insights into how RNF20 regulates CM polarity and the postnatal gene program.
Collapse
Affiliation(s)
- Chia-Yeh Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Yao-Ming Chang
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Hsin-Yi Tseng
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Yen-Ling Shih
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Hsiao-Hui Yeh
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - You-Rou Liao
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Han-Hsuan Tang
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Chia-Ling Hsu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Chien-Chang Chen
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Yu-Ting Yan
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan.
| | - Cheng-Fu Kao
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan.
| |
Collapse
|
28
|
Yu W, Gao H, Hu T, Tan X, Liu Y, Liu H, He S, Chen Z, Guo S, Huang J. Insulin-like growth factor binding protein 2: a core biomarker of left ventricular dysfunction in dilated cardiomyopathy. Hereditas 2023; 160:36. [PMID: 37904201 PMCID: PMC10617082 DOI: 10.1186/s41065-023-00298-5] [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: 06/05/2023] [Accepted: 09/18/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND RNA modifications, especially N6-methyladenosine, N1-methyladenosine and 5-methylcytosine, play an important role in the progression of cardiovascular disease. However, its regulatory function in dilated cardiomyopathy (DCM) remains to be undefined. METHODS In the study, key RNA modification regulators (RMRs) were screened by three machine learning models. Subsequently, a risk prediction model for DCM was developed and validated based on these important genes, and the diagnostic efficiency of these genes was assessed. Meanwhile, the relevance of these genes to clinical traits was explored. In both animal models and human subjects, the gene with the strongest connection was confirmed. The expression patterns of important genes were investigated using single-cell analysis. RESULTS A total of 4 key RMRs were identified. The risk prediction models were constructed basing on these genes which showed a good accuracy and sensitivity in both the training and test set. Correlation analysis showed that insulin-like growth factor binding protein 2 (IGFBP2) had the highest correlation with left ventricular ejection fraction (LVEF) (R = -0.49, P = 0.00039). Further validation expression level of IGFBP2 indicated that this gene was significantly upregulated in DCM animal models and patients, and correlation analysis validation showed a significant negative correlation between IGFBP2 and LVEF (R = -0.87; P = 6*10-5). Single-cell analysis revealed that this gene was mainly expressed in endothelial cells. CONCLUSION In conclusion, IGFBP2 is an important biomarker of left ventricular dysfunction in DCM. Future clinical applications could possibly use it as a possible therapeutic target.
Collapse
Affiliation(s)
- Wei Yu
- Department of Cardiology, The Yongchuan Hospital of Chongqing Medical University, Chongqing, China
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongli Gao
- Department of Cardiology, The Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Tianyang Hu
- Precision Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xingling Tan
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiheng Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongli Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Siming He
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zijun Chen
- Department of Cardiology, The Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Sheng Guo
- Department of Cardiology, The People's Hospital of Rongchang District, Chongqing, China.
| | - Jing Huang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
29
|
Zhang S, Yang Y, Lv X, Liu W, Zhu S, Wang Y, Xu H. Unraveling the Intricate Roles of Exosomes in Cardiovascular Diseases: A Comprehensive Review of Physiological Significance and Pathological Implications. Int J Mol Sci 2023; 24:15677. [PMID: 37958661 PMCID: PMC10650316 DOI: 10.3390/ijms242115677] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/21/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Exosomes, as potent intercellular communication tools, have garnered significant attention due to their unique cargo-carrying capabilities, which enable them to influence diverse physiological and pathological functions. Extensive research has illuminated the biogenesis, secretion, and functions of exosomes. These vesicles are secreted by cells in different states, exerting either protective or harmful biological functions. Emerging evidence highlights their role in cardiovascular disease (CVD) by mediating comprehensive interactions among diverse cell types. This review delves into the significant impacts of exosomes on CVD under stress and disease conditions, including coronary artery disease (CAD), myocardial infarction, heart failure, and other cardiomyopathies. Focusing on the cellular signaling and mechanisms, we explore how exosomes mediate multifaceted interactions, particularly contributing to endothelial dysfunction, oxidative stress, and apoptosis in CVD pathogenesis. Additionally, exosomes show great promise as biomarkers, reflecting differential expressions of NcRNAs (miRNAs, lncRNAs, and circRNAs), and as therapeutic carriers for targeted CVD treatment. However, the specific regulatory mechanisms governing exosomes in CVD remain incomplete, necessitating further exploration of their characteristics and roles in various CVD-related contexts. This comprehensive review aims to provide novel insights into the biological implications of exosomes in CVD and offer innovative perspectives on the diagnosis and treatment of CVD.
Collapse
Affiliation(s)
| | | | | | | | | | - Ying Wang
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China; (S.Z.); (Y.Y.); (X.L.); (W.L.); (S.Z.)
| | - Hongfei Xu
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China; (S.Z.); (Y.Y.); (X.L.); (W.L.); (S.Z.)
| |
Collapse
|
30
|
Benko A, Webster TJ. How to fix a broken heart-designing biofunctional cues for effective, environmentally-friendly cardiac tissue engineering. Front Chem 2023; 11:1267018. [PMID: 37901157 PMCID: PMC10602933 DOI: 10.3389/fchem.2023.1267018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/04/2023] [Indexed: 10/31/2023] Open
Abstract
Cardiovascular diseases bear strong socioeconomic and ecological impact on the worldwide healthcare system. A large consumption of goods, use of polymer-based cardiovascular biomaterials, and long hospitalization times add up to an extensive carbon footprint on the environment often turning out to be ineffective at healing such cardiovascular diseases. On the other hand, cardiac cell toxicity is among the most severe but common side effect of drugs used to treat numerous diseases from COVID-19 to diabetes, often resulting in the withdrawal of such pharmaceuticals from the market. Currently, most patients that have suffered from cardiovascular disease will never fully recover. All of these factors further contribute to the extensive negative toll pharmaceutical, biotechnological, and biomedical companies have on the environment. Hence, there is a dire need to develop new environmentally-friendly strategies that on the one hand would promise cardiac tissue regeneration after damage and on the other hand would offer solutions for the fast screening of drugs to ensure that they do not cause cardiovascular toxicity. Importantly, both require one thing-a mature, functioning cardiac tissue that can be fabricated in a fast, reliable, and repeatable manner from environmentally friendly biomaterials in the lab. This is not an easy task to complete as numerous approaches have been undertaken, separately and combined, to achieve it. This review gathers such strategies and provides insights into which succeed or fail and what is needed for the field of environmentally-friendly cardiac tissue engineering to prosper.
Collapse
Affiliation(s)
| | - Thomas J. Webster
- Department of Biomedical Engineering, Hebei University of Technology, Tianjin, China
- School of Engineering, Saveetha University, Chennai, India
- Program in Materials Science, UFPI, Teresina, Brazil
| |
Collapse
|
31
|
Li Y, Johnson JP, Yang Y, Yu D, Kubo H, Berretta RM, Wang T, Zhang X, Foster M, Yu J, Tilley DG, Houser SR, Chen X. Effects of maternal hypothyroidism on postnatal cardiomyocyte proliferation and cardiac disease responses of the progeny. Am J Physiol Heart Circ Physiol 2023; 325:H702-H719. [PMID: 37539452 PMCID: PMC10659327 DOI: 10.1152/ajpheart.00320.2023] [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: 05/31/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/05/2023]
Abstract
Maternal hypothyroidism (MH) could adversely affect the cardiac disease responses of the progeny. This study tested the hypothesis that MH reduces early postnatal cardiomyocyte (CM) proliferation so that the adult heart of MH progeny has a smaller number of larger cardiac myocytes, which imparts adverse cardiac disease responses following injury. Thyroidectomy (TX) was used to establish MH. The progeny from mice that underwent sham or TX surgery were termed Ctrl (control) or MH (maternal hypothyroidism) progeny, respectively. MH progeny had similar heart weight (HW) to body weight (BW) ratios and larger CM size consistent with fewer CMs at postnatal day 60 (P60) compared with Ctrl (control) progeny. MH progeny had lower numbers of EdU+, Ki67+, and phosphorylated histone H3 (PH3)+ CMs, which suggests they had a decreased CM proliferation in the postnatal timeframe. RNA-seq data showed that genes related to DNA replication were downregulated in P5 MH hearts, including bone morphogenetic protein 10 (Bmp10). Both in vivo and in vitro studies showed Bmp10 treatment increased CM proliferation. After transverse aortic constriction (TAC), the MH progeny had more severe cardiac pathological remodeling compared with the Ctrl progeny. Thyroid hormone (T4) treatment for MH mothers preserved their progeny's postnatal CM proliferation capacity and prevented excessive pathological remodeling after TAC. Our results suggest that CM proliferation during early postnatal development was significantly reduced in MH progeny, resulting in fewer CMs with hypertrophy in adulthood. These changes were associated with more severe cardiac disease responses after pressure overload.NEW & NOTEWORTHY Our study shows that compared with Ctrl (control) progeny, the adult progeny of mothers who have MH (MH progeny) had fewer CMs. This reduction of CM numbers was associated with decreased postnatal CM proliferation. Gene expression studies showed a reduced expression of Bmp10 in MH progeny. Bmp10 has been linked to myocyte proliferation. In vivo and in vitro studies showed that Bmp10 treatment of MH progeny and their myocytes could increase CM proliferation. Differences in CM number and size in adult hearts of MH progeny were linked to more severe cardiac structural and functional remodeling after pressure overload. T4 (synthetic thyroxine) treatment of MH mothers during their pregnancy, prevented the reduction in CM number in their progeny and the adverse response to disease stress.
Collapse
Affiliation(s)
- Yijia Li
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Jaslyn P Johnson
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Yijun Yang
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Daohai Yu
- Department of Biomedical Education and Data Science, Center for Biostatistics and Epidemiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Hajime Kubo
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Remus M Berretta
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Tao Wang
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Xiaoying Zhang
- Department of Cardiovascular Sciences, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Cardiovascular Research Center, Philadelphia, Pennsylvania, United States
| | - Michael Foster
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Jun Yu
- Department of Cardiovascular Sciences, Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Cardiovascular Research Center, Philadelphia, Pennsylvania, United States
| | - Douglas G Tilley
- Department of Cardiovascular Sciences, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Cardiovascular Research Center, Philadelphia, Pennsylvania, United States
| | - Steven R Houser
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Xiongwen Chen
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, People's Republic of China
| |
Collapse
|
32
|
Droste P, Wong DWL, Hohl M, von Stillfried S, Klinkhammer BM, Boor P. Semiautomated pipeline for quantitative analysis of heart histopathology. J Transl Med 2023; 21:666. [PMID: 37752535 PMCID: PMC10523682 DOI: 10.1186/s12967-023-04544-2] [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: 05/26/2023] [Accepted: 09/19/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND Heart diseases are among the leading causes of death worldwide, many of which lead to pathological cardiomyocyte hypertrophy and capillary rarefaction in both patients and animal models, the quantification of which is both technically challenging and highly time-consuming. Here we developed a semiautomated pipeline for quantification of the size of cardiomyocytes and capillary density in cardiac histology, termed HeartJ, by generating macros in ImageJ, a broadly used, open-source, Java-based software. METHODS We have used modified Gomori silver staining, which is easy to perform and digitize in high throughput, or Fluorescein-labeled lectin staining. The latter can be easily combined with other stainings, allowing additional quantitative analysis on the same section, e.g., the size of cardiomyocyte nuclei, capillary density, or single-cardiomyocyte protein expression. We validated the pipeline in a mouse model of cardiac hypertrophy induced by transverse aortic constriction, and in autopsy samples of patients with and without aortic stenosis. RESULTS In both animals and humans, HeartJ-based histology quantification revealed significant hypertrophy of cardiomyocytes reflecting other parameters of hypertrophy and rarefaction of microvasculature and enabling the analysis of protein expression in individual cardiomyocytes. The analysis also revealed that murine and human cardiomyocytes had similar diameters in health and extent of hypertrophy in disease confirming the translatability of our murine cardiac hypertrophy model. HeartJ enables a rapid analysis that would not be feasible by manual methods. The pipeline has little hardware requirements and is freely available. CONCLUSIONS In summary, our analysis pipeline can facilitate effective and objective quantitative histological analyses in preclinical and clinical heart samples.
Collapse
Affiliation(s)
- Patrick Droste
- LaBooratory of Nephropathology, Institute of Pathology, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Division of Nephrology and Clinical Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Dickson W L Wong
- LaBooratory of Nephropathology, Institute of Pathology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Mathias Hohl
- Department of Internal Medicine III, University Hospital, Saarland University, Homburg, Germany
| | - Saskia von Stillfried
- LaBooratory of Nephropathology, Institute of Pathology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Barbara M Klinkhammer
- LaBooratory of Nephropathology, Institute of Pathology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Peter Boor
- LaBooratory of Nephropathology, Institute of Pathology, Medical Faculty, RWTH Aachen University, Aachen, Germany.
- Division of Nephrology and Clinical Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
33
|
Velichkova G, Dobreva G. Human pluripotent stem cell-based models of heart development and disease. Cells Dev 2023; 175:203857. [PMID: 37257755 DOI: 10.1016/j.cdev.2023.203857] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/16/2023] [Accepted: 05/25/2023] [Indexed: 06/02/2023]
Abstract
The heart is a complex organ composed of distinct cell types, such as cardiomyocytes, cardiac fibroblasts, endothelial cells, smooth muscle cells, neuronal cells and immune cells. All these cell types contribute to the structural, electrical and mechanical properties of the heart. Genetic manipulation and lineage tracing studies in mice have been instrumental in gaining critical insights into the networks regulating cardiac cell lineage specification, cell fate and plasticity. Such knowledge has been of fundamental importance for the development of efficient protocols for the directed differentiation of pluripotent stem cells (PSCs) in highly specialized cardiac cell types. In this review, we summarize the evolution and current advances in protocols for cardiac subtype specification, maturation, and assembly in cardiac microtissues and organoids.
Collapse
Affiliation(s)
- Gabriel Velichkova
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gergana Dobreva
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; German Centre for Cardiovascular Research (DZHK), Germany.
| |
Collapse
|
34
|
Watanabe M, Yano T, Sato T, Umetsu A, Higashide M, Furuhashi M, Ohguro H. mTOR Inhibitors Modulate the Physical Properties of 3D Spheroids Derived from H9c2 Cells. Int J Mol Sci 2023; 24:11459. [PMID: 37511214 PMCID: PMC10380298 DOI: 10.3390/ijms241411459] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/03/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
To establish an appropriate in vitro model for the local environment of cardiomyocytes, three-dimensional (3D) spheroids derived from H9c2 cardiomyoblasts were prepared, and their morphological, biophysical phase contrast and biochemical characteristics were evaluated. The 3D H9c2 spheroids were successfully obtained, the sizes of the spheroids decreased, and they became stiffer during 3-4 days. In contrast to the cell multiplication that occurs in conventional 2D planar cell cultures, the 3D H9c2 spheroids developed into a more mature form without any cell multiplication being detected. qPCR analyses of the 3D H9c2 spheroids indicated that the production of collagen4 (COL4) and fibronectin (FN), connexin43 (CX43), β-catenin, N-cadherin, STAT3, and HIF1 molecules had increased and that the production of COL6 and α-smooth muscle actin (α-SMA) molecules had decreased as compared to 2D cultured cells. In addition, treatment with rapamycin (Rapa), an mTOR complex (mTORC) 1 inhibitor, and Torin 1, an mTORC1/2 inhibitor, resulted in significantly decreased cell densities of the 2D cultured H9c2 cells, but the size and stiffness of the H9c2 cells within the 3D spheroids were reduced with the gene expressions of several of the above several factors being reduced. The metabolic responses to mTOR modulators were also different between the 2D and 3D cultures. These results suggest that as unique aspects of the local environments of the 3D spheroids, the spontaneous expression of GJ-related molecules and hypoxia within the core may be associated with their maturation, suggesting that this may become a useful in vitro model that replicates the local environment of cardiomyocytes.
Collapse
Affiliation(s)
- Megumi Watanabe
- Department of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (M.W.); (A.U.); (M.H.)
| | - Toshiyuki Yano
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (T.Y.); (T.S.); (M.F.)
| | - Tatsuya Sato
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (T.Y.); (T.S.); (M.F.)
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Araya Umetsu
- Department of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (M.W.); (A.U.); (M.H.)
| | - Megumi Higashide
- Department of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (M.W.); (A.U.); (M.H.)
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (T.Y.); (T.S.); (M.F.)
| | - Hiroshi Ohguro
- Department of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (M.W.); (A.U.); (M.H.)
| |
Collapse
|
35
|
Mathis K, Kohon AI, Black S, Meckes B. Light-Controlled Cell-Cell Assembly Using Photocaged Oligonucleotides. ACS MATERIALS AU 2023; 3:386-393. [PMID: 38090125 PMCID: PMC10347689 DOI: 10.1021/acsmaterialsau.3c00020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/08/2023] [Accepted: 05/08/2023] [Indexed: 09/29/2024]
Abstract
The interactions between heterogeneous cell populations play important roles in dictating various cell behaviors. Cell-cell contact mediates communication through the exchange of signaling molecules, electrical coupling, and direct membrane-linked ligand-receptor interactions. In vitro culturing of multiple cell types with control over their specific arrangement is difficult, especially in three-dimensional (3D) systems. While techniques that allow one to control the arrangement of cells and direct contact between different cell types have been developed that expand upon simple co-culture methods, specific control over heterojunctions that form between cells is not easily accomplished with current methods, such as 3D cell-printing. In this article, DNA-mediated cell interactions are combined with cell-compatible photolithographic approaches to control cell assembly. Specifically, cells are coated with oligonucleotides containing DNA nucleobases that are protected with photocleavable moieties; this coating facilitated light-controlled cell assembly when these cells were mixed with cells coated with complementary oligonucleotides. By combining this technology with digital micromirror devices mounted on a microscope, selective activation of specific cell populations for interactions with other cells was achieved. Importantly, this technique is rapid and uses non-UV light sources. Taken together, this technique opens new pathways for on-demand programming of complex cell structures.
Collapse
Affiliation(s)
- Katelyn Mathis
- Department
of Biomedical Engineering, University of
North Texas, 3940 N Elm Street, Denton, Texas 76207, United States
- BioDiscovery
Institute, University of North Texas, 1155 Union Circle, Denton, Texas 76203, United States
| | - Afia Ibnat Kohon
- Department
of Biomedical Engineering, University of
North Texas, 3940 N Elm Street, Denton, Texas 76207, United States
- BioDiscovery
Institute, University of North Texas, 1155 Union Circle, Denton, Texas 76203, United States
| | - Stephen Black
- Department
of Biomedical Engineering, University of
North Texas, 3940 N Elm Street, Denton, Texas 76207, United States
- BioDiscovery
Institute, University of North Texas, 1155 Union Circle, Denton, Texas 76203, United States
| | - Brian Meckes
- Department
of Biomedical Engineering, University of
North Texas, 3940 N Elm Street, Denton, Texas 76207, United States
- BioDiscovery
Institute, University of North Texas, 1155 Union Circle, Denton, Texas 76203, United States
| |
Collapse
|
36
|
Hom LM, Sun S, Campbell J, Liu P, Culbert S, Murphy IM, Schafer ZT. A role for fibroblast-derived SASP factors in the activation of pyroptotic cell death in mammary epithelial cells. J Biol Chem 2023; 299:104922. [PMID: 37321449 PMCID: PMC10404679 DOI: 10.1016/j.jbc.2023.104922] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/17/2023] [Accepted: 06/01/2023] [Indexed: 06/17/2023] Open
Abstract
In normal tissue homeostasis, bidirectional communication between different cell types can shape numerous biological outcomes. Many studies have documented instances of reciprocal communication between fibroblasts and cancer cells that functionally change cancer cell behavior. However, less is known about how these heterotypic interactions shape epithelial cell function in the absence of oncogenic transformation. Furthermore, fibroblasts are prone to undergo senescence, which is typified by an irreversible cell cycle arrest. Senescent fibroblasts are also known to secrete various cytokines into the extracellular space; a phenomenon that is termed the senescence-associated secretory phenotype (SASP). While the role of fibroblast-derived SASP factors on cancer cells has been well studied, the impact of these factors on normal epithelial cells remains poorly understood. We discovered that treatment of normal mammary epithelial cells with conditioned media from senescent fibroblasts (SASP CM) results in a caspase-dependent cell death. This capacity of SASP CM to cause cell death is maintained across multiple senescence-inducing stimuli. However, the activation of oncogenic signaling in mammary epithelial cells mitigates the ability of SASP CM to induce cell death. Despite the reliance of this cell death on caspase activation, we discovered that SASP CM does not cause cell death by the extrinsic or intrinsic apoptotic pathway. Instead, these cells die by an NLRP3, caspase-1, and gasdermin D-dependent induction of pyroptosis. Taken together, our findings reveal that senescent fibroblasts can cause pyroptosis in neighboring mammary epithelial cells, which has implications for therapeutic strategies that perturb the behavior of senescent cells.
Collapse
Affiliation(s)
- Lisa M Hom
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Seunghoon Sun
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Jamie Campbell
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Pinyan Liu
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Shannon Culbert
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Ireland M Murphy
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Zachary T Schafer
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA.
| |
Collapse
|
37
|
Restan Perez M, da Silva VA, Cortez PE, Joddar B, Willerth SM. 3D-bioprinted cardiac tissues and their potential for disease modeling. JOURNAL OF 3D PRINTING IN MEDICINE 2023; 7:10.2217/3dp-2022-0023. [PMID: 38250545 PMCID: PMC10798787 DOI: 10.2217/3dp-2022-0023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Heart diseases cause over 17.9 million total deaths globally, making them the leading source of mortality. The aim of this review is to describe the characteristic mechanical, chemical and cellular properties of human cardiac tissue and how these properties can be mimicked in 3D bioprinted tissues. Furthermore, the authors review how current healthy cardiac models are being 3D bioprinted using extrusion-, laser- and inkjet-based printers. The review then discusses the pathologies of cardiac diseases and how bioprinting could be used to fabricate models to study these diseases and potentially find new drug targets for such diseases. Finally, the challenges and future directions of cardiac disease modeling using 3D bioprinting techniques are explored.
Collapse
Affiliation(s)
| | - Victor Alisson da Silva
- Department of Mechanical Engineering, University of Victoria, 3800 Finnerty Road, Victoria, BC, V8W 2Y2, Canada
| | - Polette Esmeralda Cortez
- Department of Metallurgical, Materials & Biomedical Engineering, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, USA
| | - Binata Joddar
- Department of Metallurgical, Materials & Biomedical Engineering, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, USA
| | - Stephanie Michelle Willerth
- Axolotl Biosciences, 3800 Finnerty Road, Victoria, BC, V8W 2Y2, Canada
- Department of Mechanical Engineering, University of Victoria, 3800 Finnerty Road, Victoria, BC, V8W 2Y2, Canada
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Road, Victoria, BC, V8W 2Y2, Canada
- Centre for Advanced Materials & Technology, University of Victoria, 3800 Finnerty Road, Victoria, BC, V8W 2Y2, Canada
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| |
Collapse
|
38
|
Spinetti G, Mutoli M, Greco S, Riccio F, Ben-Aicha S, Kenneweg F, Jusic A, de Gonzalo-Calvo D, Nossent AY, Novella S, Kararigas G, Thum T, Emanueli C, Devaux Y, Martelli F. Cardiovascular complications of diabetes: role of non-coding RNAs in the crosstalk between immune and cardiovascular systems. Cardiovasc Diabetol 2023; 22:122. [PMID: 37226245 PMCID: PMC10206598 DOI: 10.1186/s12933-023-01842-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/25/2023] [Indexed: 05/26/2023] Open
Abstract
Diabetes mellitus, a group of metabolic disorders characterized by high levels of blood glucose caused by insulin defect or impairment, is a major risk factor for cardiovascular diseases and related mortality. Patients with diabetes experience a state of chronic or intermittent hyperglycemia resulting in damage to the vasculature, leading to micro- and macro-vascular diseases. These conditions are associated with low-grade chronic inflammation and accelerated atherosclerosis. Several classes of leukocytes have been implicated in diabetic cardiovascular impairment. Although the molecular pathways through which diabetes elicits an inflammatory response have attracted significant attention, how they contribute to altering cardiovascular homeostasis is still incompletely understood. In this respect, non-coding RNAs (ncRNAs) are a still largely under-investigated class of transcripts that may play a fundamental role. This review article gathers the current knowledge on the function of ncRNAs in the crosstalk between immune and cardiovascular cells in the context of diabetic complications, highlighting the influence of biological sex in such mechanisms and exploring the potential role of ncRNAs as biomarkers and targets for treatments. The discussion closes by offering an overview of the ncRNAs involved in the increased cardiovascular risk suffered by patients with diabetes facing Sars-CoV-2 infection.
Collapse
Affiliation(s)
- Gaia Spinetti
- Laboratory of Cardiovascular Pathophysiology and Regenerative Medicine, IRCCS MultiMedica, Milan, Italy.
| | - Martina Mutoli
- Laboratory of Cardiovascular Pathophysiology and Regenerative Medicine, IRCCS MultiMedica, Milan, Italy
| | - Simona Greco
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Milan, Italy
| | - Federica Riccio
- Laboratory of Cardiovascular Pathophysiology and Regenerative Medicine, IRCCS MultiMedica, Milan, Italy
| | - Soumaya Ben-Aicha
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Franziska Kenneweg
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | | | - David de Gonzalo-Calvo
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
| | - Anne Yaël Nossent
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Susana Novella
- Department of Physiology, University of Valencia - INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Georgios Kararigas
- Department of Physiology, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Costanza Emanueli
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Milan, Italy.
| |
Collapse
|
39
|
Abstract
From the onset of the pandemic, evidence of cardiac involvement in acute COVID-19 abounded. Cardiac presentations ranged from arrhythmias to ischemia, myopericarditis/myocarditis, ventricular dysfunction to acute heart failure, and even cardiogenic shock. Elevated serum cardiac troponin levels were prevalent among hospitalized patients with COVID-19; the higher the magnitude of troponin elevation, the greater the COVID-19 illness severity and in-hospital death risk. Whether these consequences were due to direct SARS-CoV-2 infection of cardiac cells or secondary to inflammatory responses steered early cardiac autopsy studies. SARS-CoV-2 was reportedly detected in endothelial cells, cardiac myocytes, and within the extracellular space. However, findings were inconsistent and different methodologies had their limitations. Initial autopsy reports suggested that SARS-CoV-2 myocarditis was common, setting off studies to find and phenotype inflammatory infiltrates in the heart. Nonetheless, subsequent studies rarely detected myocarditis. Microthrombi, cardiomyocyte necrosis, and inflammatory infiltrates without cardiomyocyte damage were much more common. In vitro and ex vivo experimental platforms have assessed the cellular tropism of SARS-CoV-2 and elucidated mechanisms of viral entry into and replication within cardiac cells. Data point to pericytes as the primary target of SARS-CoV-2 in the heart. Infection of pericytes can account for the observed pericyte and endothelial cell death, innate immune response, and immunothrombosis commonly observed in COVID-19 hearts. These processes are bidirectional and synergistic, rendering a definitive order of events elusive. Single-cell/nucleus analyses of COVID-19 myocardial tissue and isolated cardiac cells have provided granular data about the cellular composition and cell type-specific transcriptomic signatures of COVID-19 and microthrombi-positive COVID-19 hearts. Still, much remains unknown and more in vivo studies are needed. This review seeks to provide an overview of the current understanding of COVID-19 cardiac pathophysiology. Cell type-specific mechanisms and the studies that provided such insights will be highlighted. Given the unprecedented pace of COVID-19 research, more mechanistic details are sure to emerge since the writing of this review. Importantly, our current knowledge offers significant clues about the cardiac pathophysiology of long COVID-19, the increased postrecovery risk of cardiac events, and thus, the future landscape of cardiovascular disease.
Collapse
Affiliation(s)
- Emily J Tsai
- Division of Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY (E.J.T.)
| | - Daniela Cˇiháková
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD (D.C.)
| | | |
Collapse
|
40
|
Belanger K, Koppes AN, Koppes RA. Impact of Non-Muscle Cells on Excitation-Contraction Coupling in the Heart and the Importance of In Vitro Models. Adv Biol (Weinh) 2023; 7:e2200117. [PMID: 36216583 DOI: 10.1002/adbi.202200117] [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: 04/20/2022] [Revised: 08/07/2022] [Indexed: 05/13/2023]
Abstract
Excitation-coupling (ECC) is paramount for coordinated contraction to maintain sufficient cardiac output. The study of ECC regulation has primarily been limited to cardiomyocytes (CMs), which conduct voltage waves via calcium fluxes from one cell to another, eliciting contraction of the atria followed by the ventricles. CMs rapidly transmit ionic flux via gap junction proteins, predominantly connexin 43. While the expression of connexin isoforms has been identified in each of the individual cell populations comprising the heart, the formation of gap junctions with nonmuscle cells (i.e., macrophages and Schwann cells) has gained new attention. Evaluating nonmuscle contributions to ECC in vivo or in situ remains difficult and necessitates the development of simple, yet biomimetic in vitro models to better understand and prevent physiological dysfunction. Standard 2D cell culture often consists of homogenous cell populations and lacks the dynamic mechanical environment of native tissue, confounding the phenotypic and proteomic makeup of these highly mechanosensitive cell populations in prolonged culture conditions. This review will highlight the recent developments and the importance of new microphysiological systems to better understand the complex regulation of ECC in cardiac tissue.
Collapse
Affiliation(s)
- Kirstie Belanger
- Department of Bioengineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Abigail N Koppes
- Department of Bioengineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
- Department of Biology, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| |
Collapse
|
41
|
Tan Y, Lu T, Chen Y, Witman N, Yan B, Yang L, Liu M, Gong Y, Ai X, Luo R, Wang H, Wang W, Fu W. Engineering a conduction-consistent cardiac patch with graphene oxide modified butterfly wings and human pluripotent stem cell-derived cardiomyocytes. Bioeng Transl Med 2023; 8:e10522. [PMID: 37206241 PMCID: PMC10189447 DOI: 10.1002/btm2.10522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 03/12/2023] [Accepted: 03/29/2023] [Indexed: 05/21/2023] Open
Abstract
Engineering a conduction-consistent cardiac patch has direct implications to biomedical research. However, there is difficulty in obtaining and maintaining a system that allows researchers to study physiologically relevant cardiac development, maturation, and drug screening due to the issues around inconsistent contractions of cardiomyocytes. Butterfly wings have special nanostructures arranged in parallel, which could help generate the alignment of cardiomyocytes to better mimic the natural heart tissue structure. Here, we construct a conduction-consistent human cardiac muscle patch by assembling human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) on graphene oxide (GO) modified butterfly wings. We also show this system functions as a versatile model to study human cardiomyogenesis by assembling human induced pluripotent stem cell-derived cardiac progenitor cells (hiPSC-CPCs) on the GO modified butterfly wings. The GO modified butterfly wing platform facilitated the parallel orientation of hiPSC-CMs, enhanced relative maturation as well as improved conduction consistency of the cardiomyocytes. In addition, GO modified butterfly wings enhanced the proliferation and maturation characteristics of the hiPSC-CPCs. In accordance with data obtained from RNA-sequencing and gene signatures, assembling hiPSC-CPCs on GO modified butterfly wings stimulated the differentiation of the progenitors into relatively mature hiPSC-CMs. These characteristics and capabilities of GO modified butterfly wings make them an ideal platform for heart research and drug screening.
Collapse
Affiliation(s)
- Yao Tan
- Institute of Pediatric Translational MedicineShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Tingting Lu
- Institute of Pediatric Translational MedicineShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Ying Chen
- Institute of Pediatric Translational MedicineShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Nevin Witman
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| | - Bingqian Yan
- Institute of Pediatric Translational MedicineShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Li Yang
- Department of AnesthesiologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Minglu Liu
- Department of Pediatric Cardiothoracic SurgeryShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Yiqi Gong
- Department of Pediatric Cardiothoracic SurgeryShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Xuefeng Ai
- Department of Pediatric Cardiothoracic SurgeryShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Runjiao Luo
- Department of Pediatric Cardiothoracic SurgeryShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Huijing Wang
- Institute of Pediatric Translational MedicineShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Wei Wang
- Department of Pediatric Cardiothoracic SurgeryShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Wei Fu
- Institute of Pediatric Translational MedicineShanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Tissue EngineeringShanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
42
|
Yang X, Cheng K, Wang LY, Jiang JG. The role of endothelial cell in cardiac hypertrophy: Focusing on angiogenesis and intercellular crosstalk. Biomed Pharmacother 2023; 163:114799. [PMID: 37121147 DOI: 10.1016/j.biopha.2023.114799] [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: 03/01/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/02/2023] Open
Abstract
Cardiac hypertrophy is characterized by cardiac structural remodeling, fibrosis, microvascular rarefaction, and chronic inflammation. The heart is structurally organized by different cell types, including cardiomyocytes, fibroblasts, endothelial cells, and immune cells. These cells highly interact with each other by a number of paracrine or autocrine factors. Cell-cell communication is indispensable for cardiac development, but also plays a vital role in regulating cardiac response to damage. Although cardiomyocytes and fibroblasts are deemed as key regulators of hypertrophic stimulation, other cells, including endothelial cells, also exert important effects on cardiac hypertrophy. More particularly, endothelial cells are the most abundant cells in the heart, which make up the basic structure of blood vessels and are widespread around other cells in the heart, implicating the great and inbuilt advantage of intercellular crosstalk. Cardiac microvascular plexuses are essential for transport of liquids, nutrients, molecules and cells within the heart. Meanwhile, endothelial cell-mediated paracrine signals have multiple positive or negative influences on cardiac hypertrophy. However, a comprehensive discussion of these influences and consequences is required. This review aims to summarize the basic function of endothelial cells in angiogenesis, with an emphasis on angiogenic molecules under hypertrophic conditions. The secondary objective of the research is to fully discuss the key molecules involved in the intercellular crosstalk and the endothelial cell-mediated protective or detrimental effects on other cardiac cells. This review provides a more comprehensive understanding of the overall role of endothelial cells in cardiac hypertrophy and guides the therapeutic approaches and drug development of cardiac hypertrophy.
Collapse
Affiliation(s)
- Xing Yang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430000, China
| | - Kun Cheng
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China
| | - Lu-Yun Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430000, China.
| | - Jian-Gang Jiang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430000, China.
| |
Collapse
|
43
|
Lu Y, Chen Q, An L. SPADE: Spatial Deconvolution for Domain Specific Cell-type Estimation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.14.536924. [PMID: 37131788 PMCID: PMC10153127 DOI: 10.1101/2023.04.14.536924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The advent of spatial transcriptomics technology has allowed for the acquisition of gene expression profiles with multi-cellular resolution in a spatially resolved manner, presenting a new milestone in the field of genomics. However, the aggregate gene expression from heterogeneous cell types obtained by these technologies poses a significant challenge for a comprehensive delineation of cell type-specific spatial patterns. Here, we propose SPADE (SPAtial DEconvolution), an in-silico method designed to address this challenge by incorporating spatial patterns during cell type decomposition. SPADE utilizes a combination of single-cell RNA sequencing data, spatial location information, and histological information to computationally estimate the proportion of cell types present at each spatial location. In our study, we showcased the effectiveness of SPADE by conducting analyses on synthetic data. Our results indicated that SPADE was able to successfully identify cell type-specific spatial patterns that were not previously identified by existing deconvolution methods. Furthermore, we applied SPADE to a real-world dataset analyzing the developmental chicken heart, where we observed that SPADE was able to accurately capture the intricate processes of cellular differentiation and morphogenesis within the heart. Specifically, we were able to reliably estimate changes in cell type compositions over time, which is a critical aspect of understanding the underlying mechanisms of complex biological systems. These findings underscore the potential of SPADE as a valuable tool for analyzing complex biological systems and shedding light on their underlying mechanisms. Taken together, our results suggest that SPADE represents a significant advancement in the field of spatial transcriptomics, providing a powerful tool for characterizing complex spatial gene expression patterns in heterogeneous tissues.
Collapse
|
44
|
Williams CF, Hather C, Conteh JS, Zhang J, Popa RG, Owen AW, Jonas CL, Choi H, Daniel RM, Lloyd D, Porch A, George CH. Non-thermal disruption of β-adrenergic receptor-activated Ca 2+ signalling and apoptosis in human ES-derived cardiomyocytes by microwave electric fields at 2.4 GHz. Biochem Biophys Res Commun 2023; 661:89-98. [PMID: 37087803 DOI: 10.1016/j.bbrc.2023.04.038] [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: 03/30/2023] [Accepted: 04/15/2023] [Indexed: 04/25/2023]
Abstract
The ubiquity of wireless electronic-device connectivity has seen microwaves emerge as one of the fastest growing forms of electromagnetic exposure. A growing evidence-base refutes the claim that wireless technologies pose no risk to human health at current safety levels designed to limit thermal (heating) effects. The potential impact of non-thermal effects of microwave exposure, especially in electrically-excitable tissues (e.g., heart), remains controversial. We exposed human embryonic stem-cell derived cardiomyocytes (CM), under baseline and beta-adrenergic receptor (β-AR)-stimulated conditions, to microwaves at 2.4 GHz, a frequency used extensively in wireless communication (e.g., 4G, Bluetooth™ and WiFi). To control for any effect of sample heating, experiments were done in CM subjected to matched rates of direct heating or CM maintained at 37 °C. Detailed profiling of the temporal and amplitude features of Ca2+ signalling in CM under these experimental conditions was reconciled with the extent and spatial clustering of apoptosis. The data show that exposure of CM to 2.4 GHz EMF eliminated the normal Ca2+ signalling response to β-AR stimulation and provoked spatially-clustered apoptosis. This is first evidence that non-thermal effects of 2.4 GHz microwaves might have profound effects on human CM function, responsiveness to activation, and survival.
Collapse
Affiliation(s)
- Catrin F Williams
- School of Engineering, Cardiff University, Wales, UK; School of Biosciences, Cardiff University, Wales, UK
| | | | | | | | | | | | | | - Heungjae Choi
- School of Engineering, Cardiff University, Wales, UK
| | | | - David Lloyd
- School of Engineering, Cardiff University, Wales, UK; School of Biosciences, Cardiff University, Wales, UK
| | - Adrian Porch
- School of Engineering, Cardiff University, Wales, UK.
| | | |
Collapse
|
45
|
Hom LM, Sun S, Campbell J, Liu P, Culbert S, Murphy IM, Schafer ZT. A role for fibroblast-derived SASP factors in the activation of pyroptotic cell death in mammary epithelial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.21.529458. [PMID: 36865231 PMCID: PMC9980130 DOI: 10.1101/2023.02.21.529458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
In normal tissue homeostasis, bidirectional communication between different cell types can shape numerous biological outcomes. Many studies have documented instances of reciprocal communication between fibroblasts and cancer cells that functionally change cancer cell behavior. However, less is known about how these heterotypic interactions shape epithelial cell function in the absence of oncogenic transformation. Furthermore, fibroblasts are prone to undergo senescence, which is typified by an irreversible cell cycle arrest. Senescent fibroblasts are also known to secrete various cytokines into the extracellular space; a phenomenon that is termed the senescence-associated secretory phenotype (SASP). While the role of fibroblast derived SASP factors on cancer cells has been well studied, the impact of these factors on normal epithelial cells remains poorly understood. We discovered that treatment of normal mammary epithelial cells with conditioned media (CM) from senescent fibroblasts (SASP CM) results in a caspase-dependent cell death. This capacity of SASP CM to cause cell death is maintained across multiple senescence-inducing stimuli. However, the activation of oncogenic signaling in mammary epithelial cells mitigates the ability of SASP CM to induce cell death. Despite the reliance of this cell death on caspase activation, we discovered that SASP CM does not cause cell death by the extrinsic or intrinsic apoptotic pathway. Instead, these cells die by an NLRP3, caspase-1, and gasdermin D (GSDMD)-dependent induction of pyroptosis. Taken together, our findings reveal that senescent fibroblasts can cause pyroptosis in neighboring mammary epithelial cells, which has implications for therapeutic strategies that perturb the behavior of senescent cells.
Collapse
|
46
|
Patino-Guerrero A, Ponce Wong RD, Kodibagkar VD, Zhu W, Migrino RQ, Graudejus O, Nikkhah M. Development and Characterization of Isogenic Cardiac Organoids from Human-Induced Pluripotent Stem Cells Under Supplement Starvation Regimen. ACS Biomater Sci Eng 2023; 9:944-958. [PMID: 36583992 DOI: 10.1021/acsbiomaterials.2c01290] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The prevalence of cardiovascular risk factors is expected to increase the occurrence of cardiovascular diseases (CVDs) worldwide. Cardiac organoids are promising candidates for bridging the gap between in vitro experimentation and translational applications in drug development and cardiac repair due to their attractive features. Here we present the fabrication and characterization of isogenic scaffold-free cardiac organoids derived from human induced pluripotent stem cells (hiPSCs) formed under a supplement-deprivation regimen that allows for metabolic synchronization and maturation of hiPSC-derived cardiac cells. We propose the formation of coculture cardiac organoids that include hiPSC-derived cardiomyocytes and hiPSC-derived cardiac fibroblasts (hiPSC-CMs and hiPSC-CFs, respectively). The cardiac organoids were characterized through extensive morphological assessment, evaluation of cellular ultrastructures, and analysis of transcriptomic and electrophysiological profiles. The morphology and transcriptomic profile of the organoids were improved by coculture of hiPSC-CMs with hiPSC-CFs. Specifically, upregulation of Ca2+ handling-related genes, such as RYR2 and SERCA, and structure-related genes, such as TNNT2 and MYH6, was observed. Additionally, the electrophysiological characterization of the organoids under supplement deprivation shows a trend for reduced conduction velocity for coculture organoids. These studies help us gain a better understanding of the role of other isogenic cells such as hiPSC-CFs in the formation of mature cardiac organoids, along with the introduction of exogenous chemical cues, such as supplement starvation.
Collapse
Affiliation(s)
- Alejandra Patino-Guerrero
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona8528, United States
| | | | - Vikram D Kodibagkar
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona8528, United States
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Mayo Clinic Arizona, Scottsdale, Arizona85259, United States
| | - Raymond Q Migrino
- Phoenix Veterans Affairs Health Care System, Phoenix, Arizona85012, United States.,University of Arizona College of Medicine, Phoenix, Arizona85004, United States
| | - Oliver Graudejus
- BMSEED, Mesa, Arizona85201, United States.,School of Molecular Sciences, Arizona State University, Tempe, Arizona85287, United States
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona8528, United States.,Center for Personalized Diagnostics Biodesign Institute, Arizona State University, Tempe, Arizona85281, United States
| |
Collapse
|
47
|
Barajas MB, Riess ML. Current and Novel Experimental Methods in Ischemia/Reperfusion Research: From Cutting-Edge Models to Innovative Therapies. J Vis Exp 2023:10.3791/64995. [PMID: 37255424 PMCID: PMC11087941 DOI: 10.3791/64995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024] Open
Affiliation(s)
- Matthew B Barajas
- Department of Anesthesiology, Vanderbilt University Medical Center; Department of Anesthesiology, TVHS VA Medical Center;
| | - Matthias L Riess
- Department of Anesthesiology, Vanderbilt University Medical Center; Department of Anesthesiology, TVHS VA Medical Center; Department of Pharmacology, Vanderbilt University
| |
Collapse
|
48
|
Cardiac Differentiation Promotes Focal Adhesions Assembly through Vinculin Recruitment. Int J Mol Sci 2023; 24:ijms24032444. [PMID: 36768766 PMCID: PMC9916732 DOI: 10.3390/ijms24032444] [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: 12/23/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
Cells of the cardiovascular system are physiologically exposed to a variety of mechanical forces fundamental for both cardiac development and functions. In this context, forces generated by actomyosin networks and those transmitted through focal adhesion (FA) complexes represent the key regulators of cellular behaviors in terms of cytoskeleton dynamism, cell adhesion, migration, differentiation, and tissue organization. In this study, we investigated the involvement of FAs on cardiomyocyte differentiation. In particular, vinculin and focal adhesion kinase (FAK) family, which are known to be involved in cardiac differentiation, were studied. Results revealed that differentiation conditions induce an upregulation of both FAK-Tyr397 and vinculin, resulting also in the translocation to the cell membrane. Moreover, the role of mechanical stress in contractile phenotype expression was investigated by applying a uniaxial mechanical stretching (5% substrate deformation, 1 Hz frequency). Morphological evaluation revealed that the cell shape showed a spindle shape and reoriented following the stretching direction. Substrate deformation resulted also in modification of the length and the number of vinculin-positive FAs. We can, therefore, suggest that mechanotransductive pathways, activated through FAs, are highly involved in cardiomyocyte differentiation, thus confirming their role during cytoskeleton rearrangement and cardiac myofilament maturation.
Collapse
|
49
|
Arshad S, Cutting RD, Stephens DJ, Goldberg YH, Mahr C, Vyasabattu M, Abdelfattah AH, Vaidya GN. A Peek at LVADs Pumping to Recovery. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2022; 16:11795468221144352. [PMID: 36601071 PMCID: PMC9806425 DOI: 10.1177/11795468221144352] [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: 09/18/2022] [Accepted: 11/23/2022] [Indexed: 12/27/2022]
Abstract
Left ventricular assist devices (LVADs) have revolutionized the management of patients with advanced heart failure refractory to medical therapy. Current indications of LVADs include Bridge to Transplantation (BTT), Destination Therapy (DT) for long-term use, Bridge to the Decision (BTD) used as a temporary measure, and lastly Bridge to Recovery (BTR). Here, we briefly review the clinical evidence and the molecular mechanisms behind myocardial recovery following LVAD placement. We also share institutional protocols used at 2 major medical centers in the USA.
Collapse
Affiliation(s)
- Samiullah Arshad
- Internal Medicine, University of
Kentucky, Lexington, KY, USA,Samiullah Arshad, University of Kentucky,
2151 Meeting Street, Apt 5208, Lexington, KY 40509, USA.
| | - Rachel D Cutting
- Medical Student, University of New
England College of Osteopathic Medicine, Biddeford, ME, USA
| | - Daniel J Stephens
- Medical Student, University of
Washington School of Medicine, Seattle, WA, USA
| | | | - Claudius Mahr
- Advanced Heart Failure and Transplant,
University of Washington School of Medicine, Seattle, WA, USA
| | | | | | - Gaurang N Vaidya
- Advanced Heart Failure and Transplant,
University of Kentucky, Lexington, KY, USA
| |
Collapse
|
50
|
Proteomic Insights into Cardiac Fibrosis: From Pathophysiological Mechanisms to Therapeutic Opportunities. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248784. [PMID: 36557919 PMCID: PMC9781843 DOI: 10.3390/molecules27248784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Cardiac fibrosis is a common pathophysiologic process in nearly all forms of heart disease which refers to excessive deposition of extracellular matrix proteins by cardiac fibroblasts. Activated fibroblasts are the central cellular effectors in cardiac fibrosis, and fibrotic remodelling can cause several cardiac dysfunctions either by reducing the ejection fraction due to a stiffened myocardial matrix, or by impairing electric conductance. Recently, there is a rising focus on the proteomic studies of cardiac fibrosis for pathogenesis elucidation and potential biomarker mining. This paper summarizes the current knowledge of molecular mechanisms underlying cardiac fibrosis, discusses the potential of imaging and circulating biomarkers available to recognize different phenotypes of this lesion, reviews the currently available and potential future therapies that allow individualized management in reversing progressive fibrosis, as well as the recent progress on proteomic studies of cardiac fibrosis. Proteomic approaches using clinical specimens and animal models can provide the ability to track pathological changes and new insights into the mechanisms underlining cardiac fibrosis. Furthermore, spatial and cell-type resolved quantitative proteomic analysis may also serve as a minimally invasive method for diagnosing cardiac fibrosis and allowing for the initiation of prophylactic treatment.
Collapse
|