1
|
Li L, Lu M, Guo L, Zhang X, Liu Q, Zhang M, Gao J, Xu M, Lu Y, Zhang F, Li Y, Zhang R, Liu X, Pan S, Zhang X, Li Z, Chen Y, Su X, Zhang N, Guo W, Yang T, Chen J, Qin Y, Zhang Z, Cui W, Yu L, Gu Y, Yang H, Xu X, Wang J, Burns CE, Burns CG, Han K, Zhao L, Fan G, Su Y. An organ-wide spatiotemporal transcriptomic and cellular atlas of the regenerating zebrafish heart. Nat Commun 2025; 16:3716. [PMID: 40253397 PMCID: PMC12009352 DOI: 10.1038/s41467-025-59070-0] [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: 06/12/2024] [Accepted: 04/10/2025] [Indexed: 04/21/2025] Open
Abstract
Adult zebrafish robustly regenerate injured hearts through a complex orchestration of molecular and cellular activities. However, this remarkable process, which is largely non-existent in humans, remains incompletely understood. Here, we utilize integrated spatial transcriptomics (Stereo-seq) and single-cell RNA-sequencing (scRNA-seq) to generate a spatially-resolved molecular and cellular atlas of regenerating zebrafish heart across eight stages. We characterize the cascade of cardiomyocyte cell states responsible for producing regenerated myocardium and explore a potential role for tpm4a in cardiomyocyte re-differentiation. Moreover, we uncover the activation of ifrd1 and atp6ap2 genes as a unique feature of regenerative hearts. Lastly, we reconstruct a 4D "virtual regenerating heart" comprising 569,896 cells/spots derived from 36 scRNA-seq libraries and 224 Stereo-seq slices. Our comprehensive atlas serves as a valuable resource to the cardiovascular and regeneration scientific communities and their ongoing efforts to understand the molecular and cellular mechanisms underlying vertebrate heart regeneration.
Collapse
Affiliation(s)
- Lei Li
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
| | - Meina Lu
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Lidong Guo
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuejiao Zhang
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Qun Liu
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Meiling Zhang
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Junying Gao
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Mengyang Xu
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
| | - Yijian Lu
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Fang Zhang
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Yao Li
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Ruihua Zhang
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Xiawei Liu
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Shanshan Pan
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Xianghui Zhang
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Zhen Li
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Yadong Chen
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Xiaoshan Su
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Nannan Zhang
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Wenjie Guo
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Tao Yang
- China National GeneBank, BGI Research, Shenzhen, 518120, China
| | - Jing Chen
- China National GeneBank, BGI Research, Shenzhen, 518120, China
| | - Yating Qin
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | | | - Wei Cui
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Lindong Yu
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Ying Gu
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Huanming Yang
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Xun Xu
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Jianxun Wang
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Caroline E Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - C Geoffrey Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Kai Han
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China.
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark.
| | - Long Zhao
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China.
- College of Fisheries, Ocean University of China, Qingdao, 266003, China.
| | - Guangyi Fan
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China.
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China.
- BGI Research, Sanya, 572025, China.
- BGI Research, Hangzhou, 310030, China.
| | - Ying Su
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China.
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
2
|
Punde A, Rayrikar A, Maity S, Patra C. Extracellular matrix in cardiac morphogenesis, fibrosis, and regeneration. Cells Dev 2025:204023. [PMID: 40154789 DOI: 10.1016/j.cdev.2025.204023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 03/14/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
The extracellular matrix (ECM) plays a crucial role in providing structural integrity and regulating cell communication essential for organ development, homeostasis, and regeneration, including hearts. Evidence indicates that disruptions in the spatiotemporal expression or alterations in ECM components lead to cardiac malformations, including a wide range of congenital heart diseases (CHDs). Furthermore, research on injured hearts across various vertebrate species, some of which show effective regeneration while others experience irreversible fibrosis, underscores the significance of ECM molecules in cardiac regeneration. This review presents an overview of heart development and the dynamics of ECM during cardiac morphogenesis, beginning with the formation of the contractile heart tube and advancing to the development of distinct chambers separated by valves to facilitate unidirectional blood flow. Furthermore, we discuss research emphasizing the multifaceted roles of secreted molecules in mediating fibrosis and regeneration following myocardial injury.
Collapse
Affiliation(s)
- Ashwini Punde
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra, 411004, India
| | - Amey Rayrikar
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra, 411004, India
| | - Shreya Maity
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra, 411004, India
| | - Chinmoy Patra
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra, 411004, India.
| |
Collapse
|
3
|
Rao A, Russell A, Segura-Bermudez J, Franz C, Dockery R, Blatnik A, Panten J, Zevallos M, McNulty C, Pietrzak M, Goldman JA. A cardiac transcriptional enhancer is repurposed during regeneration to activate an anti-proliferative program. Development 2025; 152:DEV204458. [PMID: 39803985 PMCID: PMC11883283 DOI: 10.1242/dev.204458] [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: 10/15/2024] [Accepted: 12/17/2024] [Indexed: 02/18/2025]
Abstract
Zebrafish have a high capacity to regenerate their hearts. Several studies have surveyed transcriptional enhancers to understand how gene expression is controlled during heart regeneration. We have identified REN (the runx1 enhancer) that, during regeneration, regulates the expression of the nearby runx1 gene. We show that runx1 mRNA is reduced with deletion of REN (ΔREN), and cardiomyocyte proliferation is enhanced in ΔREN mutants only during regeneration. Interestingly, in uninjured hearts, ΔREN mutants have reduced expression of adamts1, a nearby gene that encodes a Collagen protease. This results in excess Collagen within cardiac valves of uninjured hearts. The ΔREN Collagen phenotype is rescued by an allele with Δrunx1 mutations, suggesting that in uninjured hearts REN regulates adamts1 independently of runx1. Taken together, this suggests that REN is rewired from adamts1 in uninjured hearts to stimulate runx1 transcription during regeneration. Our data point to a previously unappreciated mechanism for gene regulation during zebrafish heart regeneration. We report that an enhancer is rewired from expression in a distal cardiac domain to activate a different gene in regenerating tissue.
Collapse
Affiliation(s)
- Anupama Rao
- Department of Biological Chemistry and Pharmacology, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - Andrew Russell
- Department of Biological Chemistry and Pharmacology, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - Jose Segura-Bermudez
- Department of Biological Chemistry and Pharmacology, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - Charles Franz
- Department of Biological Chemistry and Pharmacology, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - Rejenae Dockery
- Department of Biological Chemistry and Pharmacology, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - Anton Blatnik
- Department of Biological Chemistry and Pharmacology, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - Jacob Panten
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Mateo Zevallos
- Department of Biological Chemistry and Pharmacology, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - Carson McNulty
- Department of Biological Chemistry and Pharmacology, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - Maciej Pietrzak
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - Joseph Aaron Goldman
- Department of Biological Chemistry and Pharmacology, The Ohio State University Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
4
|
Tam E, Ouimet M, Sweeney G. Cardioprotective Effects of Adiponectin-Stimulated Autophagy. J Lipid Atheroscler 2025; 14:40-53. [PMID: 39911962 PMCID: PMC11791421 DOI: 10.12997/jla.2025.14.1.40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/11/2024] [Accepted: 09/19/2024] [Indexed: 02/07/2025] Open
Abstract
Cardiovascular diseases (CVDs), including heart failure, pose a significant economic and health burden worldwide. Current treatment strategies for heart failure are greatly limited, in that they mainly mitigate symptoms or delay further progression. In contrast, therapies aimed at proactively preventing the onset of heart failure could greatly improve outcomes. Adiponectin is an adipocyte-derived hormone that confers an array of cardioprotective effects. It exerts anti-inflammatory effects, improves metabolic function, mitigates endothelial cell dysfunction, and reduce cardiomyocyte cell death. Furthermore, it has gained increasing attention for its ability to activate autophagy, a conserved cellular pathway that facilitates the degradation and recycling of cell components. The disruption of autophagy has been linked to CVDs including heart failure. Additionally, growing evidence also points to specific forms of autophagy, namely mitophagy and lipophagy, as crucial adaptive responses in protection against CVDs. The protective effects of adiponectin, autophagy, mitophagy, and lipophagy against CVDs along with potential therapeutic implications will be discussed.
Collapse
Affiliation(s)
- Eddie Tam
- Department of Biology, York University, Toronto, ON, Canada
| | - Mireille Ouimet
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, Canada
| |
Collapse
|
5
|
Toporcer T, Grendel T, Špaková I, Blichárová A, Verbóová Ľ, Benetinová Z, Čižmárová B, Rabajdová M, Toporcerová S. An In Vivo Model of Estrogen Supplementation Concerning the Expression of Ca 2+-Dependent Exchangers and Mortality, Vitality and Survival After Myocardial Infarction in Ovariectomized Rats. J Cardiovasc Dev Dis 2024; 11:352. [PMID: 39590195 PMCID: PMC11595027 DOI: 10.3390/jcdd11110352] [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: 09/22/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Ischemic-reperfusion damage of cardiomyocytes due to myocardial infarction (MI) often leads to the death of an individual. Premenopausal women have been observed to have a significantly lower risk of cardiovascular disease (CVD) than men of the same age. In menopausal women, this trend is significantly reversed, and the risk of CVD increases up to 10-fold. Estrogens affect the development and function of the heart muscle, and as they decrease, the risk and poor prognosis of CVD increase. This study is focused on the effects of estrogen supplementation on morbidity, vitality, and NCX1 expression after MI on a model system. METHODS In this study, female Sprague Dawley rats (n = 58), which were divided into three experimental groups (NN-control group, non-supplemented; OVX-N-ovariectomized, non-supplemented; OVX-S-ovariectomized, supplemented), received left thoracotomy in the fourth intercostal space. The left anterior descendent coronary artery was ligated 2 mm from its origin with an 8.0 suture. An immunohistological analysis as well as an RT-PCR analysis of NCX1 expression were performed. RESULTS A higher survival rate was recorded in the OVX-N group (86%) in comparison with the OVX-S group (53%) (p < 0.05). In addition, higher NCX1 expression 7 days/14 days after MI in the OVX-S group in comparison with the NN and OVX-N (p < 0.001 and p < 0.05) groups was recorded. Seven days after MI, a significantly higher expression (p < 0.005) of mRNA NCX1 in the OVX-N group was also recorded in comparison with the NN group. CONCLUSIONS This study provides a comprehensive description of the effect of estrogen supplementation on NCX1 expression and overall vitality in ovariectomized rats that survived MI.
Collapse
Affiliation(s)
- Tomáš Toporcer
- Department of Heart Surgery, East Slovak Institute of Cardiovascular Disease and Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia;
| | - Tomáš Grendel
- Department of Anesthesiology and Intensive Medicine, East Slovak Institute of Cardiovascular Disease and Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia
| | - Ivana Špaková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia (B.Č.); (M.R.)
| | - Alžbeta Blichárová
- Department of Pathology, Louis Pasteur University Hospital and Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (A.B.); (Ľ.V.); (Z.B.)
| | - Ľudmila Verbóová
- Department of Pathology, Louis Pasteur University Hospital and Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (A.B.); (Ľ.V.); (Z.B.)
| | - Zuzana Benetinová
- Department of Pathology, Louis Pasteur University Hospital and Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (A.B.); (Ľ.V.); (Z.B.)
| | - Beata Čižmárová
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia (B.Č.); (M.R.)
| | - Miroslava Rabajdová
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia (B.Č.); (M.R.)
| | - Silvia Toporcerová
- Department of Gynecology and Obstetrics, Faculty of Medicine, Pavol Jozef Šafárik University and Gyncare, 040 11 Košice, Slovakia
| |
Collapse
|
6
|
Li Z, Zhang M, Wang Y, Li Y, Zhu YZ. A Novel Human Amniotic Membrane Suspension Improves the Therapeutic Effect of Mesenchymal Stem Cells on Myocardial Infarction in Rats. ADVANCED NANOBIOMED RESEARCH 2024; 4. [DOI: 10.1002/anbr.202400084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Mesenchymal stem cell (MSC) therapy aids cardiac repair and regeneration, but the low rate of MSC survival and engulfment in the infarcted heart remains a major obstacle for routine clinical application. Here, an injectable suspension of human acellular amniotic membrane (HAAM) that may serve as synergistic cell delivery vehicle for the treatment of myocardial infarction (MI) by improving MSC homing and survival is developed. The results demonstrate that compared with MSC transplantation alone, HAAM‐loaded MSCs have higher survival and engraftment rates in infarcted tissue, alleviated hypoxia‐induced myocardial damage, achieved higher improvements in cardiac function, promoted angiogenesis, and reduced myocardial fibrosis. In addition, HAAM‐loaded MSCs increase N‐cadherin levels and thereby enhance the efficacy of MSCs in treating MI. This study provides a new approach for MSC‐based cardiac repair and regeneration.
Collapse
Affiliation(s)
- Zhaoyi Li
- Pharmacy, Faculty of Medicine & State Key Laboratory of Quality Research in Chinese Medicine Macau University of Science and Technology Macau 999078 China
| | - Meirong Zhang
- Huaxia (Qingdao) Biotech Co., Ltd. Qingdao 266000 China
- Huaxia Regenerative Medicine Institute for Human Materials Langfang 065000 China
| | - Yi Wang
- Huaxia (Qingdao) Biotech Co., Ltd. Qingdao 266000 China
- Huaxia Regenerative Medicine Institute for Human Materials Langfang 065000 China
| | - Yijia Li
- Huaxia (Qingdao) Biotech Co., Ltd. Qingdao 266000 China
- Huaxia Regenerative Medicine Institute for Human Materials Langfang 065000 China
| | - Yi Zhun Zhu
- Pharmacy, Faculty of Medicine & State Key Laboratory of Quality Research in Chinese Medicine Macau University of Science and Technology Macau 999078 China
- Laboratory of Drug Discovery from Natural Resources and Industrialization, School of Pharmacy Macau University of Science and Technology Macau 999078 China
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy Fudan University Shanghai 201203 China
| |
Collapse
|
7
|
Sauvage E, Matta J, Dang CT, Fan J, Cruzado G, Cicoira F, Merle G. Electroconductive cardiac patch based on bioactive PEDOT:PSS hydrogels. J Biomed Mater Res A 2024; 112:1817-1826. [PMID: 38689450 DOI: 10.1002/jbm.a.37729] [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: 12/26/2023] [Revised: 04/13/2024] [Accepted: 04/22/2024] [Indexed: 05/02/2024]
Abstract
Engineering cardiac implants for treating myocardial infarction (MI) has advanced, but challenges persist in mimicking the structural properties and variability of cardiac tissues using traditional bioconstructs and conventional engineering methods. This study introduces a synthetic patch with a bioactive surface designed to swiftly restore functionality to the damaged myocardium. The patch combines a composite, soft, and conductive hydrogel-based on (3,4-ethylenedioxythiophene):polystyrene-sulfonate (PEDOT:PSS) and polyvinyl alcohol (PVA). This cardiac patch exhibits a reasonably high electrical conductivity (40 S/cm) and a stretchability up to 50% of its original length. Our findings reveal its resilience to 10% cyclic stretching at 1 Hz with no loss of conductivity over time. To mediate a strong cell-scaffold adhesion, we biofunctionalize the hydrogel with a N-cadherin mimic peptide, providing the cardiac patch with a bioactive surface. This modification promote increased adherence and proliferation of cardiac fibroblasts (CFbs) while effectively mitigating the formation of bacterial biofilm, particularly against Staphylococcus aureus, a common pathogen responsible for surgical site infections (SSIs). Our study demonstrates the successful development of a structurally validated cardiac patch possessing the desired mechanical, electrical, and biofunctional attributes for effective cardiac recovery. Consequently, this research holds significant promise in alleviating the burden imposed by myocardial infarctions.
Collapse
Affiliation(s)
- Erwan Sauvage
- Department of Chemical Engineering, Polytechnique Montréal, Montréal, Quebec, Canada
| | - Justin Matta
- Department of Experimental Surgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Cat-Thy Dang
- Department of Chemical Engineering, Polytechnique Montréal, Montréal, Quebec, Canada
| | - Jiaxin Fan
- Department of Chemical Engineering, Polytechnique Montréal, Montréal, Quebec, Canada
| | - Graziele Cruzado
- Department of Chemical Engineering, Polytechnique Montréal, Montréal, Quebec, Canada
| | - Fabio Cicoira
- Department of Chemical Engineering, Polytechnique Montréal, Montréal, Quebec, Canada
| | - Géraldine Merle
- Department of Chemical Engineering, Polytechnique Montréal, Montréal, Quebec, Canada
| |
Collapse
|
8
|
Liang J, He X, Wang Y. Cardiomyocyte proliferation and regeneration in congenital heart disease. PEDIATRIC DISCOVERY 2024; 2:e2501. [PMID: 39308981 PMCID: PMC11412308 DOI: 10.1002/pdi3.2501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/25/2024] [Indexed: 09/25/2024]
Abstract
Despite advances in prenatal screening and a notable decrease in mortality rates, congenital heart disease (CHD) remains the most prevalent congenital disorder in newborns globally. Current therapeutic surgical approaches face challenges due to the significant rise in complications and disabilities. Emerging cardiac regenerative therapies offer promising adjuncts for CHD treatment. One novel avenue involves investigating methods to stimulate cardiomyocyte proliferation. However, the mechanism of altered cardiomyocyte proliferation in CHD is not fully understood, and there are few feasible approaches to stimulate cardiomyocyte cell cycling for optimal healing in CHD patients. In this review, we explore recent progress in understanding genetic and epigenetic mechanisms underlying defective cardiomyocyte proliferation in CHD from development through birth. Targeting cell cycle pathways shows promise for enhancing cardiomyocyte cytokinesis, division, and regeneration to repair heart defects. Advancements in human disease modeling techniques, CRISPR-based genome and epigenome editing, and next-generation sequencing technologies will expedite the exploration of abnormal machinery governing cardiomyocyte differentiation, proliferation, and maturation across diverse genetic backgrounds of CHD. Ongoing studies on screening drugs that regulate cell cycling are poised to translate this nascent technology of enhancing cardiomyocyte proliferation into a new therapeutic paradigm for CHD surgical interventions.
Collapse
Affiliation(s)
- Jialiang Liang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Xingyu He
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
9
|
Marín LO, Montoya Y, Bustamante J. Biological Evaluation of Thermosensitive Hydrogels of Chitosan/Hydrolyzed Collagen/β-GP in an In Vitro Model of Induced Cardiac Ischemia. Polymers (Basel) 2024; 16:2206. [PMID: 39125232 PMCID: PMC11314826 DOI: 10.3390/polym16152206] [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: 11/21/2023] [Revised: 12/07/2023] [Accepted: 12/15/2023] [Indexed: 08/12/2024] Open
Abstract
Ischemic events can culminate in acute myocardial infarction, which is generated by irreversible cardiac lesions that cannot be restored due to the limited regenerative capacity of the heart. Cardiac cell therapy aims to replace injured or necrotic cells with healthy and functional cells. Tissue engineering and cardiovascular regenerative medicine propose therapeutic alternatives using biomaterials that mimic the native extracellular environment and improve cellular and tissue functionality. This investigation evaluates the effect of thermosensitive hydrogels, and murine fetal ventricular cardiomyocytes encapsulated in thermosensitive hydrogels, on the contractile function of cardiomyocyte regeneration during an ischemic event. Chitosan and hydrolyzed collagen thermosensitive hydrogels were developed, and they were physically and chemically characterized. Likewise, their biocompatibility was evaluated through cytotoxicity assays by MTT, LDH, and their hemolytic capacity. The hydrogels, and cells inside the hydrogels, were used as an intervention for primary cardiomyocytes under hypoxic conditions to determine the restoration of the contractile capacity by measuring intracellular calcium levels and the expressions of binding proteins, such as a-actinin and connexin 43. These results evidence the potential of natural thermosensitive hydrogels to restore the bioelectrical functionality of ischemic cardiomyocytes.
Collapse
Affiliation(s)
- Lina Orozco Marín
- Tissue Engineering and Cardiovascular Prosthetics Line, Cardiovascular Dynamics Group, Bioengineering Center, Universidad Pontificia Bolivariana, Medellín 050004, Colombia; (L.O.M.); (J.B.)
| | - Yuliet Montoya
- Tissue Engineering and Cardiovascular Prosthetics Line, Cardiovascular Dynamics Group, Bioengineering Center, Universidad Pontificia Bolivariana, Medellín 050004, Colombia; (L.O.M.); (J.B.)
- Working Committee of Cardiovascular Bioengineering, Colombian Society of Cardiology and Cardiovascular Surgery, Bogotá 1013, Colombia
| | - John Bustamante
- Tissue Engineering and Cardiovascular Prosthetics Line, Cardiovascular Dynamics Group, Bioengineering Center, Universidad Pontificia Bolivariana, Medellín 050004, Colombia; (L.O.M.); (J.B.)
- Working Committee of Cardiovascular Bioengineering, Colombian Society of Cardiology and Cardiovascular Surgery, Bogotá 1013, Colombia
| |
Collapse
|
10
|
Hassan S, Rezaei Z, Luna E, Yilmaz-Aykut D, Lee MC, Perea AM, Jamaiyar A, Bassous N, Hirano M, Tourk FM, Choi C, Becker M, Yazdi I, Fan K, Avila-Ramirez A, Ge D, Abdi R, Fisch S, Leijten J, Feinberg MW, Mandal BB, Liao R, Shin SR. Injectable Self-Oxygenating Cardio-Protective and Tissue Adhesive Silk-Based Hydrogel for Alleviating Ischemia After Mi Injury. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2312261. [PMID: 38733225 PMCID: PMC11309903 DOI: 10.1002/smll.202312261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/23/2024] [Indexed: 05/13/2024]
Abstract
Myocardial infarction (MI) is a significant cardiovascular disease that restricts blood flow, resulting in massive cell death and leading to stiff and noncontractile fibrotic scar tissue formation. Recently, sustained oxygen release in the MI area has shown regeneration ability; however, improving its therapeutic efficiency for regenerative medicine remains challenging. Here, a combinatorial strategy for cardiac repair by developing cardioprotective and oxygenating hybrid hydrogels that locally sustain the release of stromal cell-derived factor-1 alpha (SDF) and oxygen for simultaneous activation of neovascularization at the infarct area is presented. A sustained release of oxygen and SDF from injectable, mechanically robust, and tissue-adhesive silk-based hybrid hydrogels is achieved. Enhanced endothelialization under normoxia and anoxia is observed. Furthermore, there is a marked improvement in vascularization that leads to an increment in cardiomyocyte survival by ≈30% and a reduction of the fibrotic scar formation in an MI animal rodent model. Improved left ventricular systolic and diastolic functions by ≈10% and 20%, respectively, with a ≈25% higher ejection fraction on day 7 are also observed. Therefore, local delivery of therapeutic oxygenating and cardioprotective hydrogels demonstrates beneficial effects on cardiac functional recovery for reparative therapy.
Collapse
Affiliation(s)
- Shabir Hassan
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
| | - Zahra Rezaei
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
| | - Eder Luna
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
| | - Dilara Yilmaz-Aykut
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Chemical Engineering, Faculty of Engineering, Istanbul University-Cerrahpaşa, 34320 Istanbul, Turkey
| | - Myung Chul Lee
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
| | - Ana Marie Perea
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
| | - Anurag Jamaiyar
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Nicole Bassous
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
| | - Minoru Hirano
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
- Future Vehicle Research Department, Toyota Research Institute North America, Toyota Motor North America, Inc., 1555 Woodridge Ave., Ann Arbor, Michigan 48105, USA
| | - Fatima Mumtaza Tourk
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Mechanical Engineering, Northeastern University, Boston, MA 02115, USA
| | - Cholong Choi
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
| | - Malin Becker
- Department of Developmental BioEngineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands
| | - Iman Yazdi
- School of Arts and Sciences, Regis College, 235 Wellesley Street, Weston, MA 02493, USA
| | - Kai Fan
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
- BoYu Intelligent Health Innovation Laboratory, Hangzhou 311121, China
| | - Alan Avila-Ramirez
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
- Division of Biological and Environmental Science Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - David Ge
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital / Harvard Medical School, Boston, MA 02115, USA
| | - Sudeshna Fisch
- Cardiovascular Physiology Core, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Jeroen Leijten
- Department of Developmental BioEngineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands
| | - Mark W. Feinberg
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Biman B. Mandal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Ronglih Liao
- School of Medicine, Stanford University, California 94305-5101, USA
- Stanford Amyloid Center, Stanford University, California 94305-5101, USA
| | - Su Ryon Shin
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
| |
Collapse
|
11
|
Koukorava C, Ahmed K, Almaghrabi S, Pointon A, Haddrick M, Cross MJ. Anticancer drugs and cardiotoxicity: the role of cardiomyocyte and non-cardiomyocyte cells. Front Cardiovasc Med 2024; 11:1372817. [PMID: 39081368 PMCID: PMC11287221 DOI: 10.3389/fcvm.2024.1372817] [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: 01/18/2024] [Accepted: 05/31/2024] [Indexed: 08/02/2024] Open
Abstract
Cardiotoxicity can be defined as "chemically induced heart disease", which can occur with many different drug classes treating a range of diseases. It is the primary cause of drug attrition during pre-clinical development and withdrawal from the market. Drug induced cardiovascular toxicity can result from both functional effects with alteration of the contractile and electrical regulation in the heart and structural changes with morphological changes to cardiomyocytes and other cardiac cells. These adverse effects result in conditions such as arrhythmia or a more serious reduction in left ventricular ejection fraction (LVEF), which can lead to heart failure and death. Anticancer drugs can adversely affect cardiomyocyte function as well as cardiac fibroblasts and cardiac endothelial cells, interfering in autocrine and paracrine signalling between these cell types and ultimately altering cardiac cellular homeostasis. This review aims to highlight potential toxicity mechanisms involving cardiomyocytes and non-cardiomyocyte cells by first introducing the physiological roles of these cells within the myocardium and secondly, identifying the physiological pathways perturbed by anticancer drugs in these cells.
Collapse
Affiliation(s)
- Chrysa Koukorava
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Katie Ahmed
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Shrouq Almaghrabi
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Amy Pointon
- Safety Sciences, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | | | - Michael J. Cross
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Liverpool Centre for Cardiovascular Science, Liverpool, United Kingdom
| |
Collapse
|
12
|
Seth J, Sharma S, Leong CJ, Vaibhav V, Nelson P, Shokravi A, Luo Y, Shirvani D, Laksman Z. The Use of Hematopoietic Stem Cells for Heart Failure: A Systematic Review. Int J Mol Sci 2024; 25:6634. [PMID: 38928341 PMCID: PMC11204149 DOI: 10.3390/ijms25126634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/04/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
The purpose of this review is to summarize the current understanding of the therapeutic effect of stem cell-based therapies, including hematopoietic stem cells, for the treatment of ischemic heart damage. Following PRISMA guidelines, we conducted electronic searches in MEDLINE, and EMBASE. We screened 592 studies, and included RCTs, observational studies, and cohort studies that examined the effect of hematopoietic stem cell therapy in adult patients with heart failure. Studies that involved pediatric patients, mesenchymal stem cell therapy, and non-heart failure (HF) studies were excluded from our review. Out of the 592 studies, 7 studies met our inclusion criteria. Overall, administration of hematopoietic stem cells (via intracoronary or myocardial infarct) led to positive cardiac outcomes such as improvements in pathological left-ventricular remodeling, perfusion following acute myocardial infarction, and NYHA symptom class. Additionally, combined death, rehospitalization for heart failure, and infarction were significantly lower in patients treated with bone marrow-derived hematopoietic stem cells. Our review demonstrates that hematopoietic stem cell administration can lead to positive cardiac outcomes for HF patients. Future studies should aim to increase female representation and non-ischemic HF patients.
Collapse
Affiliation(s)
- Jayant Seth
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; (J.S.); (S.S.); (C.J.L.); (A.S.); (Y.L.); (D.S.)
| | - Sohat Sharma
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; (J.S.); (S.S.); (C.J.L.); (A.S.); (Y.L.); (D.S.)
| | - Cameron J. Leong
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; (J.S.); (S.S.); (C.J.L.); (A.S.); (Y.L.); (D.S.)
| | - Venkat Vaibhav
- Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; (V.V.); (P.N.)
| | - Pierce Nelson
- Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; (V.V.); (P.N.)
| | - Arveen Shokravi
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; (J.S.); (S.S.); (C.J.L.); (A.S.); (Y.L.); (D.S.)
| | - Yuchen Luo
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; (J.S.); (S.S.); (C.J.L.); (A.S.); (Y.L.); (D.S.)
| | - Daniel Shirvani
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; (J.S.); (S.S.); (C.J.L.); (A.S.); (Y.L.); (D.S.)
| | - Zachary Laksman
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; (J.S.); (S.S.); (C.J.L.); (A.S.); (Y.L.); (D.S.)
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC V6Z 1Y6, Canada
| |
Collapse
|
13
|
Zeinelabdeen Y, Abaza T, Yasser MB, Elemam NM, Youness RA. MIAT LncRNA: A multifunctional key player in non-oncological pathological conditions. Noncoding RNA Res 2024; 9:447-462. [PMID: 38511054 PMCID: PMC10950597 DOI: 10.1016/j.ncrna.2024.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/27/2023] [Accepted: 01/14/2024] [Indexed: 03/22/2024] Open
Abstract
The discovery of non-coding RNAs (ncRNAs) has unveiled a wide range of transcripts that do not encode proteins but play key roles in several cellular and molecular processes. Long noncoding RNAs (lncRNAs) are specific class of ncRNAs that are longer than 200 nucleotides and have gained significant attention due to their diverse mechanisms of action and potential involvement in various pathological conditions. In the current review, the authors focus on the role of lncRNAs, specifically highlighting the Myocardial Infarction Associated Transcript (MIAT), in non-oncological context. MIAT is a nuclear lncRNA that has been directly linked to myocardial infarction and is reported to control post-transcriptional processes as a competitive endogenous RNA (ceRNA) molecule. It interacts with microRNAs (miRNAs), thereby limiting the translation and expression of their respective target messenger RNA (mRNA) and regulating protein expression. Yet, MIAT has been implicated in other numerous pathological conditions such as other cardiovascular diseases, autoimmune disease, neurodegenerative diseases, metabolic diseases, and many others. In this review, the authors emphasize that MIAT exhibits distinct expression patterns and functions across different pathological conditions and is emerging as potential diagnostic, prognostic, and therapeutic agent. Additionally, the authors highlight the regulatory role of MIAT and shed light on the involvement of lncRNAs and specifically MIAT in various non-oncological pathological conditions.
Collapse
Affiliation(s)
- Yousra Zeinelabdeen
- Molecular Genetics Research Team, Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU), Cairo, 11835, Egypt
- Faculty of Medical Sciences/UMCG, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713 AV, the Netherlands
| | - Tasneem Abaza
- Molecular Genetics Research Team, Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU), Cairo, 11835, Egypt
- Biotechnology and Biomolecular Biochemistry Program, Faculty of Science, Cairo University, Cairo, Egypt
| | - Montaser Bellah Yasser
- Bioinformatics Group, Center for Informatics Sciences (CIS), School of Information Technology and Computer Science (ITCS), Nile University, Giza, Egypt
| | - Noha M. Elemam
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Rana A. Youness
- Molecular Genetics Research Team, Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU), Cairo, 11835, Egypt
| |
Collapse
|
14
|
Salar Amoli M, Ma Z, Nakada Y, Fukuda K, Zhang J, Serpooshan V. Editorial: Bioengineering and biotechnology approaches in cardiovascular regenerative medicine, volume II. Front Bioeng Biotechnol 2024; 12:1380646. [PMID: 38433821 PMCID: PMC10904635 DOI: 10.3389/fbioe.2024.1380646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 03/05/2024] Open
Affiliation(s)
- Mehdi Salar Amoli
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA, United States
| | - Zhen Ma
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, United States
- BioInspired Syracuse Institute for Material and Living Systems, Syracuse University, Syracuse, NY, United States
| | - Yuji Nakada
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Vahid Serpooshan
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Children’s Healthcare of Atlanta, Atlanta, GA, United States
| |
Collapse
|
15
|
Guo A, Zhang S, Yang R, Sui C. [Not Available]. Mater Today Bio 2024; 24:100939. [PMID: 38249436 PMCID: PMC10797197 DOI: 10.1016/j.mtbio.2023.100939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
Gelatin methacrylate (GelMA) hydrogels have gained significant traction in diverse tissue engineering applications through the utilization of 3D printing technology. As an artificial hydrogel possessing remarkable processability, GelMA has emerged as a pioneering material in the advancement of tissue engineering due to its exceptional biocompatibility and degradability. The integration of 3D printing technology facilitates the precise arrangement of cells and hydrogel materials, thereby enabling the creation of in vitro models that simulate artificial tissues suitable for transplantation. Consequently, the potential applications of GelMA in tissue engineering are further expanded. In tissue engineering applications, the mechanical properties of GelMA are often modified to overcome the hydrogel material's inherent mechanical strength limitations. This review provides a comprehensive overview of recent advancements in enhancing the mechanical properties of GelMA at the monomer, micron, and nano scales. Additionally, the diverse applications of GelMA in soft tissue engineering via 3D printing are emphasized. Furthermore, the potential opportunities and obstacles that GelMA may encounter in the field of tissue engineering are discussed. It is our contention that through ongoing technological progress, GelMA hydrogels with enhanced mechanical strength can be successfully fabricated, leading to the production of superior biological scaffolds with increased efficacy for tissue engineering purposes.
Collapse
Affiliation(s)
- Ao Guo
- Department of Trauma and Pediatric Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 231200, China
| | - Shengting Zhang
- Department of Trauma and Pediatric Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 231200, China
| | - Runhuai Yang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, China
| | - Cong Sui
- Department of Trauma and Pediatric Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 231200, China
| |
Collapse
|
16
|
Navab R, Haward R, Chacko J, Haward R. Platelet-Rich Plasma for Heart Cell Regeneration Post-myocardial Infarction: A Propitious Therapeutic Approach. Cureus 2024; 16:e51951. [PMID: 38333505 PMCID: PMC10852202 DOI: 10.7759/cureus.51951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2024] [Indexed: 02/10/2024] Open
Abstract
Globally, one of the primary factors leading to death is cardiovascular disorders, specifically coronary artery disease, which leads to myocardial infarction (MI). This article investigates the potential of platelet-rich plasma (PRP) therapy for regenerating cardiac cells following MI. We look into the pathophysiology of MI, current treatment methods, and the heart's limited ability to heal itself. This is done to see if PRP could help the heart heal faster, reduce the size of the infarct, and stop scar tissue from forming. We analyze the production procedure of PRP, its composition of growth factors, and its utilization in many medical domains. The ways that PRP helps the heart heal are also being looked into. This includes how it affects inflammation, oxidative stress, angiogenesis, and cell proliferation. Although we recognize the existing constraints, we meticulously take into account issues such as standardization, therapeutic variance, and potential harmful effects. This study highlights the importance of comprehensive guidelines, continuous research, and enhanced clinical applications to fully harness the potential of platelet-rich plasma in the regeneration of cardiac cells after a heart attack.
Collapse
Affiliation(s)
- Rahul Navab
- Internal Medicine, PES Institute of Medical Sciences and Research, Kuppam, IND
| | - Raymond Haward
- Internal Medicine, Vydehi Institute of Medical Sciences and Research Centre, Bangalore, IND
| | - Joshua Chacko
- Internal Medicine, Father Muller Medical College, Mangalore, IND
| | - Rachel Haward
- Internal Medicine, KVG Medical College and Hospital, Sullia, IND
| |
Collapse
|
17
|
Huang H, Huang GN, Payumo AY. Two decades of heart regeneration research: Cardiomyocyte proliferation and beyond. WIREs Mech Dis 2024; 16:e1629. [PMID: 37700522 PMCID: PMC10840678 DOI: 10.1002/wsbm.1629] [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: 05/15/2023] [Revised: 08/03/2023] [Accepted: 08/09/2023] [Indexed: 09/14/2023]
Abstract
Interest in vertebrate cardiac regeneration has exploded over the past two decades since the discovery that adult zebrafish are capable of complete heart regeneration, contrasting the limited regenerative potential typically observed in adult mammalian hearts. Undercovering the mechanisms that both support and limit cardiac regeneration across the animal kingdom may provide unique insights in how we may unlock this capacity in adult humans. In this review, we discuss key discoveries in the heart regeneration field over the last 20 years. Initially, seminal findings revealed that pre-existing cardiomyocytes are the major source of regenerated cardiac muscle, drawing interest into the intrinsic mechanisms regulating cardiomyocyte proliferation. Moreover, recent studies have identified the importance of intercellular interactions and physiological adaptations, which highlight the vast complexity of the cardiac regenerative process. Finally, we compare strategies that have been tested to increase the regenerative capacity of the adult mammalian heart. This article is categorized under: Cardiovascular Diseases > Stem Cells and Development.
Collapse
Affiliation(s)
- Herman Huang
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192, USA
| | - Guo N. Huang
- Cardiovascular Research Institute & Department of Physiology, University of California, San Francisco, San Francisco, CA, 94158, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Alexander Y. Payumo
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192, USA
| |
Collapse
|
18
|
Cai Y, Xiong M, Xin Z, Liu C, Ren J, Yang X, Lei J, Li W, Liu F, Chu Q, Zhang Y, Yin J, Ye Y, Liu D, Fan Y, Sun S, Jing Y, Zhao Q, Zhao L, Che S, Zheng Y, Yan H, Ma S, Wang S, Izpisua Belmonte JC, Qu J, Zhang W, Liu GH. Decoding aging-dependent regenerative decline across tissues at single-cell resolution. Cell Stem Cell 2023; 30:1674-1691.e8. [PMID: 37898124 DOI: 10.1016/j.stem.2023.09.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/28/2023] [Accepted: 09/27/2023] [Indexed: 10/30/2023]
Abstract
Regeneration across tissues and organs exhibits significant variation throughout the body and undergoes a progressive decline with age. To decode the relationships between aging and regenerative capacity, we conducted a comprehensive single-cell transcriptome analysis of regeneration in eight tissues from young and aged mice. We employed diverse analytical models to study tissue regeneration and unveiled the intricate cellular and molecular mechanisms underlying the attenuated regenerative processes observed in aged tissues. Specifically, we identified compromised stem cell mobility and inadequate angiogenesis as prominent contributors to this age-associated decline in regenerative capacity. Moreover, we discovered a unique subset of Arg1+ macrophages that were activated in young tissues but suppressed in aged regenerating tissues, suggesting their important role in age-related immune response disparities during regeneration. This study provides a comprehensive single-cell resource for identifying potential targets for interventions aimed at enhancing regenerative outcomes in the aging population.
Collapse
Affiliation(s)
- Yusheng Cai
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Muzhao Xiong
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zijuan Xin
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Chengyu Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Jie Ren
- Key Laboratory of RNA Science and Engineering, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; Aging Biomarker Consortium, China
| | - Xiying Yang
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - Jinghui Lei
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Wei Li
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Feifei Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Qun Chu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yiyuan Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Jian Yin
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yanxia Ye
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Dingyi Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yanling Fan
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Shuhui Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yaobin Jing
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Qian Zhao
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Liyun Zhao
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Shanshan Che
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yandong Zheng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Haoteng Yan
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Aging Biomarker Consortium, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Aging Biomarker Consortium, China
| | | | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Aging Biomarker Consortium, China.
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Aging Biomarker Consortium, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Aging Biomarker Consortium, China.
| |
Collapse
|
19
|
Apaydin O, Altaikyzy A, Filosa A, Sawamiphak S. Alpha-1 adrenergic signaling drives cardiac regeneration via extracellular matrix remodeling transcriptional program in zebrafish macrophages. Dev Cell 2023; 58:2460-2476.e7. [PMID: 37875117 DOI: 10.1016/j.devcel.2023.09.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 06/24/2023] [Accepted: 09/29/2023] [Indexed: 10/26/2023]
Abstract
The autonomic nervous system plays a pivotal role in cardiac repair. Here, we describe the mechanistic underpinning of adrenergic signaling in fibrotic and regenerative response of the heart to be dependent on immunomodulation. A pharmacological approach identified adrenergic receptor alpha-1 as a key regulator of macrophage phenotypic diversification following myocardial damage in zebrafish. Genetic manipulation and single-cell transcriptomics showed that the receptor signals activation of an "extracellular matrix remodeling" transcriptional program in a macrophage subset, which serves as a key regulator of matrix composition and turnover. Mechanistically, adrenergic receptor alpha-1-activated macrophages determine activation of collagen-12-expressing fibroblasts, a cellular determinant of cardiac regenerative niche, through midkine-mediated paracrine crosstalk, allowing lymphatic and blood vessel growth and cardiomyocyte proliferation at the lesion site. These findings identify the mechanism of adrenergic signaling in macrophage phenotypic and functional determination and highlight the potential of neural modulation for regulation of fibrosis and coordination of myocardial regenerative response.
Collapse
Affiliation(s)
- Onur Apaydin
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Institute of Chemistry and Biochemistry, Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Akerke Altaikyzy
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Alessandro Filosa
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Suphansa Sawamiphak
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
| |
Collapse
|
20
|
Driss LB, Lian J, Walker RG, Howard JA, Thompson TB, Rubin LL, Wagers AJ, Lee RT. GDF11 and aging biology - controversies resolved and pending. THE JOURNAL OF CARDIOVASCULAR AGING 2023; 3:42. [PMID: 38235060 PMCID: PMC10793994 DOI: 10.20517/jca.2023.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Since the exogenous administration of GDF11, a TGF-ß superfamily member, was reported to have beneficial effects in some models of human disease, there have been many research studies in GDF11 biology. However, many studies have now confirmed that exogenous administration of GDF11 can improve physiology in disease models, including cardiac fibrosis, experimental stroke, and disordered metabolism. GDF11 is similar to GDF8 (also called Myostatin), differing only by 11 amino acids in their mature signaling domains. These two proteins are now known to be biochemically different both in vitro and in vivo. GDF11 is much more potent than GDF8 and induces more strongly SMAD2 phosphorylation in the myocardium compared to GDF8. GDF8 and GDF11 prodomain are only 52% identical and are cleaved by different Tolloid proteases to liberate the mature signaling domain from inhibition of the prodomain. Here, we review the state of GDF11 biology, highlighting both resolved and remaining controversies.
Collapse
Affiliation(s)
- Laura Ben Driss
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - John Lian
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Ryan G. Walker
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45267, USA
| | - James A. Howard
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Thomas B. Thompson
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Lee L. Rubin
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Amy J. Wagers
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
- Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Joslin Diabetes Center, Boston, MA 02115, USA
| | - Richard T. Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| |
Collapse
|
21
|
Parker JB, Valencia C, Akras D, DiIorio SE, Griffin MF, Longaker MT, Wan DC. Understanding Fibroblast Heterogeneity in Form and Function. Biomedicines 2023; 11:2264. [PMID: 37626760 PMCID: PMC10452440 DOI: 10.3390/biomedicines11082264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Historically believed to be a homogeneous cell type that is often overlooked, fibroblasts are more and more understood to be heterogeneous in nature. Though the mechanisms behind how fibroblasts participate in homeostasis and pathology are just beginning to be understood, these cells are believed to be highly dynamic and play key roles in fibrosis and remodeling. Focusing primarily on fibroblasts within the skin and during wound healing, we describe the field's current understanding of fibroblast heterogeneity in form and function. From differences due to embryonic origins to anatomical variations, we explore the diverse contributions that fibroblasts have in fibrosis and plasticity. Following this, we describe molecular techniques used in the field to provide deeper insights into subpopulations of fibroblasts and their varied roles in complex processes such as wound healing. Limitations to current work are also discussed, with a focus on future directions that investigators are recommended to take in order to gain a deeper understanding of fibroblast biology and to develop potential targets for translational applications in a clinical setting.
Collapse
Affiliation(s)
- Jennifer B. Parker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (M.F.G.)
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Caleb Valencia
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (M.F.G.)
| | - Deena Akras
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (M.F.G.)
| | - Sarah E. DiIorio
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (M.F.G.)
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michelle F. Griffin
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (M.F.G.)
| | - Michael T. Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (M.F.G.)
| | - Derrick C. Wan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA (M.F.G.)
| |
Collapse
|
22
|
Cosentino M, Nicoletti C, Valenti V, Schirone L, Di Nonno F, Apa L, Zouhair M, Genovese D, Madaro L, Dinarelli S, Rossi M, Del Prete Z, Sciarretta S, Frati G, Rizzuto E, Musarò A. Remodeled eX vivo muscle engineered tissue improves heart function after chronic myocardial ischemia. Sci Rep 2023; 13:10370. [PMID: 37365262 DOI: 10.1038/s41598-023-37553-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 06/23/2023] [Indexed: 06/28/2023] Open
Abstract
The adult heart displays poor reparative capacities after injury. Cell transplantation and tissue engineering approaches have emerged as possible therapeutic options. Several stem cell populations have been largely used to treat the infarcted myocardium. Nevertheless, transplanted cells displayed limited ability to establish functional connections with the host cardiomyocytes. In this study, we provide a new experimental tool, named 3D eX vivo muscle engineered tissue (X-MET), to define the contribution of mechanical stimuli in triggering functional remodeling and to rescue cardiac ischemia. We revealed that mechanical stimuli trigger a functional remodeling of the 3D skeletal muscle system toward a cardiac muscle-like structure. This was supported by molecular and functional analyses, demonstrating that remodeled X-MET expresses relevant markers of functional cardiomyocytes, compared to unstimulated and to 2D- skeletal muscle culture system. Interestingly, transplanted remodeled X-MET preserved heart function in a murine model of chronic myocardial ischemia and increased survival of transplanted injured mice. X-MET implantation resulted in repression of pro-inflammatory cytokines, induction of anti-inflammatory cytokines, and reduction in collagen deposition. Altogether, our findings indicate that biomechanical stimulation induced a cardiac functional remodeling of X-MET, which showed promising seminal results as a therapeutic product for the development of novel strategies for regenerative medicine.
Collapse
Affiliation(s)
- Marianna Cosentino
- Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14, 00161, Rome, Italy
| | - Carmine Nicoletti
- Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14, 00161, Rome, Italy
| | - Valentina Valenti
- Department of Cardiology, Ospedale Santa Maria Goretti, 04100, Latina, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Leonardo Schirone
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | | | - Ludovica Apa
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, 00184, Rome, Italy
| | - Mariam Zouhair
- Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14, 00161, Rome, Italy
| | - Desiree Genovese
- Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14, 00161, Rome, Italy
| | - Luca Madaro
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - Simone Dinarelli
- Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, 00161, Rome, Italy
| | - Marco Rossi
- Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, 00161, Rome, Italy
| | - Zaccaria Del Prete
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, 00184, Rome, Italy
| | - Sebastiano Sciarretta
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- IRCCS Neuromed, Pozzilli (IS), Italy
| | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- IRCCS Neuromed, Pozzilli (IS), Italy
| | - Emanuele Rizzuto
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, 00184, Rome, Italy
| | - Antonio Musarò
- Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14, 00161, Rome, Italy.
- Scuola Superiore di Studi Avanzati Sapienza (SSAS), Sapienza University of Rome, 00185, Rome, Italy.
| |
Collapse
|
23
|
Okamura DM, Nguyen ED, Collins SJ, Yoon K, Gere JB, Weiser-Evans MCM, Beier DR, Majesky MW. Mammalian organ regeneration in spiny mice. J Muscle Res Cell Motil 2023; 44:39-52. [PMID: 36131170 DOI: 10.1007/s10974-022-09631-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/30/2022] [Indexed: 11/26/2022]
Abstract
Fibrosis-driven solid organ failure is a major world-wide health burden with few therapeutic options. Spiny mice (genus: Acomys) are terrestrial mammals that regenerate severe skin wounds without fibrotic scars to evade predators. Recent studies have shown that spiny mice also regenerate acute ischemic and traumatic injuries to kidney, heart, spinal cord, and skeletal muscle. A common feature of this evolved wound healing response is a lack of formation of fibrotic scar tissue that degrades organ function, inhibits regeneration, and leads to organ failure. Complex tissue regeneration is an extremely rare property among mammalian species. In this article, we discuss the evidence that Acomys represents an emerging model organism that offers a unique opportunity for the biomedical community to investigate and clinically translate molecular mechanisms of scarless wound healing and regeneration of organ function in a mammalian species.
Collapse
Affiliation(s)
- Daryl M Okamura
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA
| | - Elizabeth D Nguyen
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA
| | - Sarah J Collins
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA
| | - Kevin Yoon
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA
| | - Joshua B Gere
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA
| | - Mary C M Weiser-Evans
- Department of Medicine, Division of Renal Diseases & Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - David R Beier
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA
| | - Mark W Majesky
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA.
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, 98195, USA.
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
24
|
Budharaju H, Sundaramurthi D, Sethuraman S. Efficient dual crosslinking of protein-in-polysaccharide bioink for biofabrication of cardiac tissue constructs. BIOMATERIALS ADVANCES 2023; 152:213486. [PMID: 37302210 DOI: 10.1016/j.bioadv.2023.213486] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/13/2023]
Abstract
Myocardial infarction (MI) is a lethal cardiac disease that causes most of the mortality across the world. MI is a consequence of plaque in the arterial walls of heart, which eventually result in occlusion and ischemia to the myocardial tissues due to inadequate nutrient and oxygen supply. As an efficient alternative to the existing treatment strategies for MI, 3D bioprinting has evolved as an advanced tissue fabrication technique where the cell-laden bioinks are printed layer-by-layer to create functional cardiac patches. In this study, a dual crosslinking strategy has been utilized towards 3D bioprinting of myocardial constructs by using a combination of alginate and fibrinogen. Herein, pre-crosslinking of the physically blended alginate-fibrinogen bioinks with CaCl2 enhanced the shape fidelity and printability of the printed structures. Physicochemical properties of the bioinks such as rheology, fibrin distribution, swelling ratio and degradation behaviour, were determined post-printing for only ionically crosslinked & dual crosslinked constructs and found to be ideal for bioprinting of cardiac constructs. Human ventricular cardiomyocytes (AC 16) exhibited a significant increase in cell proliferation on day 7 and 14 in AF-DMEM-20 mM CaCl2 bioink when compared to A-DMEM-20 mM CaCl2 (p < 0.05). Furthermore, myocardial patches containing neonatal ventricular rat myocytes (NVRM) showed >80 % viability and also expressed sarcomeric alpha actinin & connexin 43. These results indicate that the dual crosslinking strategy was cytocompatible and also possess the potential to be used for biofabrication of thick myocardial constructs for regenerative medicine applications.
Collapse
Affiliation(s)
- Harshavardhan Budharaju
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India
| | - Dhakshinamoorthy Sundaramurthi
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India.
| |
Collapse
|
25
|
Aziz R, Falanga M, Purenovic J, Mancini S, Lamberti P, Guida M. A Review on the Applications of Natural Biodegradable Nano Polymers in Cardiac Tissue Engineering. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:1374. [PMID: 37110959 PMCID: PMC10145986 DOI: 10.3390/nano13081374] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 06/19/2023]
Abstract
As cardiac diseases, which mostly result in heart failure, are increasing rapidly worldwide, heart transplantation seems the only solution for saving lives. However, this practice is not always possible due to several reasons, such as scarcity of donors, rejection of organs from recipient bodies, or costly medical procedures. In the framework of nanotechnology, nanomaterials greatly contribute to the development of these cardiovascular scaffolds as they provide an easy regeneration of the tissues. Currently, functional nanofibers can be used in the production of stem cells and in the regeneration of cells and tissues. The small size of nanomaterials, however, leads to changes in their chemical and physical characteristics that could alter their interaction and exposure to stem cells with cells and tissues. This article aims to review the naturally occurring biodegradable nanomaterials that are used in cardiovascular tissue engineering for the development of cardiac patches, vessels, and tissues. Moreover, this article also provides an overview of cell sources used for cardiac tissue engineering, explains the anatomy and physiology of the human heart, and explores the regeneration of cardiac cells and the nanofabrication approaches used in cardiac tissue engineering as well as scaffolds.
Collapse
Affiliation(s)
- Rabia Aziz
- Department of Information and Electrical Engineering and Applied Mathematics (DIEM), University of Salerno, 84084 Fisciano, Italy; (M.F.); (S.M.); (P.L.); (M.G.)
- Consiglio Nazionale Delle Ricerche (CNR)-Istituto Officina dei Materiali (IOM), Area Science Park Basovizza S.S. 14-Km. 163, 5-34149 Trieste, Italy
| | - Mariarosaria Falanga
- Department of Information and Electrical Engineering and Applied Mathematics (DIEM), University of Salerno, 84084 Fisciano, Italy; (M.F.); (S.M.); (P.L.); (M.G.)
| | - Jelena Purenovic
- Department of Physics and Materials, Faculty of Sciences at Cacak, University of Kragujevac, 32000 Cacak, Serbia;
| | - Simona Mancini
- Department of Information and Electrical Engineering and Applied Mathematics (DIEM), University of Salerno, 84084 Fisciano, Italy; (M.F.); (S.M.); (P.L.); (M.G.)
| | - Patrizia Lamberti
- Department of Information and Electrical Engineering and Applied Mathematics (DIEM), University of Salerno, 84084 Fisciano, Italy; (M.F.); (S.M.); (P.L.); (M.G.)
- Italian Interuniversity Research Center on Interaction between Electromagnetic Fields and Biosystems (ICEmB), Università Degli Studi di Genova, DITEN, Via all’Opera Pia 11/a, 16145 Genova, Italy
- Interdepartmental Research Centre for Nanomaterials and Nanotechnology at the University of Salerno (NanoMates), Department of Physics, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| | - Michele Guida
- Department of Information and Electrical Engineering and Applied Mathematics (DIEM), University of Salerno, 84084 Fisciano, Italy; (M.F.); (S.M.); (P.L.); (M.G.)
- Italian Interuniversity Research Center on Interaction between Electromagnetic Fields and Biosystems (ICEmB), Università Degli Studi di Genova, DITEN, Via all’Opera Pia 11/a, 16145 Genova, Italy
| |
Collapse
|
26
|
Dvinskikh L, Sparks H, Brito L, MacLeod KT, Harding SE, Dunsby C. Remote-refocusing light-sheet fluorescence microscopy enables 3D imaging of electromechanical coupling of hiPSC-derived and adult cardiomyocytes in co-culture. Sci Rep 2023; 13:3342. [PMID: 36849727 PMCID: PMC9970973 DOI: 10.1038/s41598-023-29419-w] [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: 09/19/2022] [Accepted: 02/03/2023] [Indexed: 03/01/2023] Open
Abstract
Improving cardiac function through stem-cell regenerative therapy requires functional and structural integration of the transplanted cells with the host tissue. Visualizing the electromechanical interaction between native and graft cells necessitates 3D imaging with high spatio-temporal resolution and low photo-toxicity. A custom light-sheet fluorescence microscope was used for volumetric imaging of calcium dynamics in co-cultures of adult rat left ventricle cardiomyocytes and human induced pluripotent stem cell-derived cardiomyocytes. Aberration-free remote refocus of the detection plane synchronously to the scanning of the light sheet along the detection axis enabled fast dual-channel 3D imaging at subcellular resolution without mechanical sample disturbance at up to 8 Hz over a ∼300 µm × 40 µm × 50 µm volume. The two cell types were found to undergo electrically stimulated and spontaneous synchronized calcium transients and contraction. Electromechanical coupling improved with co-culture duration, with 50% of adult-CM coupled after 24 h of co-culture, compared to 19% after 4 h (p = 0.0305). Immobilization with para-nitroblebbistatin did not prevent calcium transient synchronization, with 35% and 36% adult-CM coupled in control and treated samples respectively (p = 0.91), indicating that electrical coupling can be maintained independently of mechanotransduction.
Collapse
Affiliation(s)
- L Dvinskikh
- Department of Physics, Imperial College London, London, UK. .,National Heart and Lung Institute, Imperial College London, London, UK. .,Department of Chemistry, Imperial College London, London, UK.
| | - H Sparks
- Department of Physics, Imperial College London, London, UK
| | - L Brito
- National Heart and Lung Institute, Imperial College London, London, UK
| | - K T MacLeod
- National Heart and Lung Institute, Imperial College London, London, UK
| | - S E Harding
- National Heart and Lung Institute, Imperial College London, London, UK
| | - C Dunsby
- Department of Physics, Imperial College London, London, UK
| |
Collapse
|
27
|
Renikunta H, Chakrabarti R, Duddu S, Bhattacharya A, Chakravorty N, Shukla PC. Stem Cells and Therapies in Cardiac Regeneration. Regen Med 2023. [DOI: 10.1007/978-981-19-6008-6_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
28
|
Dewing JM, Saunders V, O’Kelly I, Wilson DI. Defining cardiac cell populations and relative cellular composition of the early fetal human heart. PLoS One 2022; 17:e0259477. [PMID: 36449524 PMCID: PMC9710754 DOI: 10.1371/journal.pone.0259477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/08/2022] [Indexed: 12/03/2022] Open
Abstract
While the adult human heart is primarily composed of cardiomyocytes, fibroblasts, endothelial and smooth muscle cells, the cellular composition during early development remains largely unknown. Reliable identification of fetal cardiac cell types using protein markers is critical to understand cardiac development and delineate the cellular composition of the developing human heart. This is the first study to use immunohistochemistry (IHC), flow cytometry and RT-PCR analyses to investigate the expression and specificity of commonly used cardiac cell markers in the early human fetal heart (8-12 post-conception weeks). The expression of previously reported protein markers for the detection of cardiomyocytes (Myosin Heavy Chain (MHC) and cardiac troponin I (cTnI), fibroblasts (DDR2, THY1, Vimentin), endothelial cells (CD31) and smooth muscle cells (α-SMA) were assessed. Two distinct populations of cTnI positive cells were identified through flow cytometry, with MHC positive cardiomyocytes showing high cTnI expression (cTnIHigh) while MHC negative non-myocytes showed lower cTnI expression (cTnILow). cTnI expression in non-myocytes was further confirmed by IHC and RT-PCR analyses, suggesting troponins are not cardiomyocyte-specific and may play distinct roles in non-muscle cells during early development. Vimentin (VIM) was expressed in cultured ventricular fibroblast populations and flow cytometry revealed VIMHigh and VIMLow cell populations in the fetal heart. MHC positive cardiomyocytes were VIMLow whilst CD31 positive endothelial cells were VIMHigh. Using markers investigated within this study, we characterised fetal human cardiac populations and estimate that 75-80% of fetal cardiac cells are cardiomyocytes and are MHC+/cTnIHigh/VIMLow, whilst non-myocytes comprise 20-25% of total cells and are MHC-/cTnILow/VIMHigh, with CD31+ endothelial cells comprising ~9% of this population. These findings show distinct differences from those reported for adult heart.
Collapse
Affiliation(s)
- Jennifer M. Dewing
- Institute for Developmental Sciences, School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- * E-mail:
| | - Vinay Saunders
- Institute for Developmental Sciences, School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Ita O’Kelly
- Institute for Developmental Sciences, School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Immunocore Ltd, Abingdon, Oxford, United Kingdom
| | - David I. Wilson
- Institute for Developmental Sciences, School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
29
|
Altered Cellular Protein Quality Control System Modulates Cardiomyocyte Function in Volume Overload-Induced Hypertrophy. Antioxidants (Basel) 2022; 11:antiox11112210. [DOI: 10.3390/antiox11112210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 10/29/2022] [Accepted: 11/03/2022] [Indexed: 11/10/2022] Open
Abstract
Volume-induced hypertrophy is one of the risk factors for cardiac morbidity and mortality. In addition, mechanical and metabolic dysfunction, aging, and cellular redox balance are also contributing factors to the disease progression. In this study, we used volume overload (VO), which was induced by an aortocaval fistula in 2-month-old male Wistar rats, and sham-operated animals served as control. Functional parameters were measured by transthoracic echocardiography at termination 4- or 8-months after VO. The animals showed hypertrophic remodeling that was accompanied by mechanical dysfunction and increased cardiomyocyte stiffness. These alterations were reversible upon treatment with glutathione. Cardiomyocyte dysfunction was associated with elevated oxidative stress markers with unchanged inflammatory signaling pathways. In addition, we observed altered phosphorylation status of small heat shock proteins 27 and 70 and diminished protease expression caspases 3 compared to the matched control group, indicating an impaired protein quality control system. Such alterations might be attributed to the increased oxidative stress as anticipated from the enhanced titin oxidation, ubiquitination, and the elevation in oxidative stress markers. Our study showed an early pathological response to VO, which manifests in cardiomyocyte mechanical dysfunction and dysregulated signaling pathways associated with enhanced oxidative stress and an impaired protein quality control system.
Collapse
|
30
|
Defining the molecular underpinnings controlling cardiomyocyte proliferation. Clin Sci (Lond) 2022; 136:911-934. [PMID: 35723259 DOI: 10.1042/cs20211180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 05/27/2022] [Accepted: 05/31/2022] [Indexed: 12/11/2022]
Abstract
Shortly after birth, mammalian cardiomyocytes (CM) exit the cell cycle and cease to proliferate. The inability of adult CM to replicate renders the heart particularly vulnerable to injury. Restoration of CM proliferation would be an attractive clinical target for regenerative therapies that can preserve contractile function and thus prevent the development of heart failure. Our review focuses on recent progress in understanding the tight regulation of signaling pathways and their downstream molecular mechanisms that underly the inability of CM to proliferate in vivo. In this review, we describe the temporal expression of cell cycle activators e.g., cyclin/Cdk complexes and their inhibitors including p16, p21, p27 and members of the retinoblastoma gene family during gestation and postnatal life. The differential impact of members of the E2f transcription factor family and microRNAs on the regulation of positive and negative cell cycle factors is discussed. This review also highlights seminal studies that identified the coordination of signaling mechanisms that can potently activate CM cell cycle re-entry including the Wnt/Ctnnb1, Hippo, Pi3K-Akt and Nrg1-Erbb2/4 pathways. We also present an up-to-date account of landmark studies analyzing the effect of various genes such as Argin, Dystrophin, Fstl1, Meis1, Pitx2 and Pkm2 that are responsible for either inhibition or activation of CM cell division. All these reports describe bona fide therapeutically targets that could guide future clinical studies toward cardiac repair.
Collapse
|
31
|
Zhang X, Zhang Q, Chen L, Cai B, Zeng M, Ou S, Chen Y, Feng Z, Chen H, Cao S, Kang K. Appropriate Exogenous Expression Stoichiometry of GATA4 as an Important Factor for Cardiac Reprogramming of Human Dermal Fibroblasts. Cell Reprogram 2022; 24:283-293. [PMID: 35762944 DOI: 10.1089/cell.2022.0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Reprogramming of human dermal fibroblasts (HDFs) into induced cardiomyocyte-like cells (iCMs) represents a promising strategy for human cardiac regeneration. Different cocktails of cardiac transcription factors can convert HDFs into iCMs, although with low efficiency and immature phenotype. Here, GATA4, MEF2C, TBX5, MESP1, and MYOCD (GMTMeMy for short) were used to reprogram HDFs by retrovirus infection. We found that the exogenous expression stoichiometry of GATA4 (GATA4 stoichiometry) significantly affected reprogramming efficiency. When 1/8 dosage of GATA4 virus (GATA4 dosage) plus MTMeMy was used, the reprogramming efficiency was obviously improved compared with average pooled virus encoding each factor, which measured, by the expression level of cardiac genes, the percentage of cardiac troponin T and alpha-cardiac myosin heavy-chain immunopositive cells and the numbers of iCMs showing calcium oscillation or beating synchronously in co-culture with mouse CMs. In addition, we prepared conditioned maintenance medium (CMM) by CM differentiation of H9 human embryonic stem cell line. We found that compared with traditional maintenance medium (TMM), CMM made iCMs show well-organized sarcomere formation and characteristic calcium oscillation wave earlier. These findings demonstrated that appropriate GATA4 stoichiometry was essential for cardiac reprogramming and some components in CMM were important for maturation of iCMs.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Qi Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lijun Chen
- Joint School of Life Science, Guangzhou Institutes of Biomedicine and Health, Chinese Academic and Sciences, Guangzhou Medical University, Guangzhou, China
| | - Baomei Cai
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Center for Cell Lineage and Atlas, Guangzhou, China
| | - Mengying Zeng
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Center for Cell Lineage and Atlas, Guangzhou, China
| | - Sihua Ou
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yating Chen
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ziyu Feng
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Center for Cell Lineage and Atlas, Guangzhou, China
| | - Huan Chen
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Center for Cell Lineage and Atlas, Guangzhou, China
| | - Shangtao Cao
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Center for Cell Lineage and Atlas, Guangzhou, China
| | - Kai Kang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
32
|
Ross Stewart KM, Walker SL, Baker AH, Riley PR, Brittan M. Hooked on heart regeneration: the zebrafish guide to recovery. Cardiovasc Res 2022; 118:1667-1679. [PMID: 34164652 PMCID: PMC9215194 DOI: 10.1093/cvr/cvab214] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/22/2021] [Indexed: 12/23/2022] Open
Abstract
While humans lack sufficient capacity to undergo cardiac regeneration following injury, zebrafish can fully recover from a range of cardiac insults. Over the past two decades, our understanding of the complexities of both the independent and co-ordinated injury responses by multiple cardiac tissues during zebrafish heart regeneration has increased exponentially. Although cardiomyocyte regeneration forms the cornerstone of the reparative process in the injured zebrafish heart, recent studies have shown that this is dependent on prior neovascularization and lymphangiogenesis, which in turn require epicardial, endocardial, and inflammatory cell signalling within an extracellular milieu that is optimized for regeneration. Indeed, it is the amalgamation of multiple regenerative systems and gene regulatory patterns that drives the much-heralded success of the adult zebrafish response to cardiac injury. Increasing evidence supports the emerging paradigm that developmental transcriptional programmes are re-activated during adult tissue regeneration, including in the heart, and the zebrafish represents an optimal model organism to explore this concept. In this review, we summarize recent advances from the zebrafish cardiovascular research community with novel insight into the mechanisms associated with endogenous cardiovascular repair and regeneration, which may be of benefit to inform future strategies for patients with cardiovascular disease.
Collapse
Affiliation(s)
- Katherine M Ross Stewart
- Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Sophie L Walker
- Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Andrew H Baker
- Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Paul R Riley
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Sherrington Rd, Oxford OX1 3PT, UK
| | - Mairi Brittan
- Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
33
|
Wiggs MP, Beaudry AG, Law ML. Cardiac Remodeling in Cancer-Induced Cachexia: Functional, Structural, and Metabolic Contributors. Cells 2022; 11:cells11121931. [PMID: 35741060 PMCID: PMC9221803 DOI: 10.3390/cells11121931] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/01/2022] [Accepted: 06/08/2022] [Indexed: 11/17/2022] Open
Abstract
Cancer cachexia is a syndrome of progressive weight loss and muscle wasting occurring in many advanced cancer patients. Cachexia significantly impairs quality of life and increases mortality. Cardiac atrophy and dysfunction have been observed in patients with cachexia, which may contribute to cachexia pathophysiology. However, relative to skeletal muscle, little research has been carried out to understand the mechanisms of cardiomyopathy in cachexia. Here, we review what is known clinically about the cardiac changes occurring in cachexia, followed by further discussion of underlying physiological and molecular mechanisms contributing to cachexia-induced cardiomyopathy. Impaired cardiac contractility and relaxation may be explained by a complex interplay of significant heart muscle atrophy and metabolic remodeling, including mitochondrial dysfunction. Because cardiac muscle has fundamental differences compared to skeletal muscle, understanding cardiac-specific effects of cachexia may bring light to unique therapeutic targets and ultimately improve clinical management for patients with cancer cachexia.
Collapse
Affiliation(s)
- Michael P. Wiggs
- Department of Health, Human Performance, and Recreation, Robbins College of Health and Human Sciences, Baylor University, Waco, TX 76706, USA; (M.P.W.); (A.G.B.)
| | - Anna G. Beaudry
- Department of Health, Human Performance, and Recreation, Robbins College of Health and Human Sciences, Baylor University, Waco, TX 76706, USA; (M.P.W.); (A.G.B.)
| | - Michelle L. Law
- Department of Human Sciences and Design, Robbins College of Health and Human Sciences, Baylor University, Waco, TX 76706, USA
- Correspondence: ; Tel.: +1-(254)-710-6003
| |
Collapse
|
34
|
Li Y, Yang M, Tan J, Shen C, Deng S, Fu X, Gao S, Li H, Zhang X, Cai W. Targeting ACSL1 promotes cardiomyocyte proliferation and cardiac regeneration. Life Sci 2022; 294:120371. [PMID: 35122795 DOI: 10.1016/j.lfs.2022.120371] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/19/2021] [Accepted: 01/28/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Neonatal hearts have considerable regenerative potential within 7 days post birth (P7), but the rate of regeneration is extremely low after P7. Interestingly, lipid metabolism increases dramatically after P7. The similarities in these age profiles suggests a possible link between cardiac regeneration and lipid metabolism. Acyl CoA synthase long chain family member 1 (ACSL1) is the key enzyme that regulates lipid metabolism. The aim of this study was to identify the role of ACSL1 in the regeneration of cardiomyocytes. METHODS AND RESULTS The uptake of fatty acids in hearts increased after P7; however, myocardial regeneration was decreased. We profiled an RNA-sequence array of hearts from mice of different ages, including E10.5 (embryonic stage)-, 3-, 7-, 21-, 30-, and 60-day-old mice, and found that the expression of ACSL1 was significantly increased after P7. By establishing ACSL1 knockdown mice with adeno-associated virus (AAV9). Then, we verified that knockdown of ACSL1 enhanced the capacity for myocardial regeneration both in mice and in primary cardiomyocytes. Indeed, ACSL1 knockdown in primary cardiomyocytes promoted the cell cycle progression from G0 to G2 phase by regulating specific factors, which may correlate with the activation of AKT by ACSL1 and withdrawal of FOXO1 from the nucleus. In vivo, knockdown of ACSL1 effectively restored cardiac function and myocardial regeneration in adult mice with myocardial infarction (MI). CONCLUSIONS ACSL1 possibly induces the loss of the myocardial regenerative potential beginning at P7 in mice, and inhibition of ACSL1 effectively promoted myocardial repair after MI in mice.
Collapse
Affiliation(s)
- Yuanlong Li
- Institute of Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Ming Yang
- Institute of Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Jing Tan
- Institute of Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Conghui Shen
- Institute of Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Shijie Deng
- Institute of Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Xinlu Fu
- Institute of Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Saifei Gao
- Institute of Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Hui Li
- Institute of Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Xiaoxue Zhang
- The Second Department of Cardiology, Guangdong Second Provincial General Hospital & Guangdong Provincial Emergency Hospital, Guangzhou 510317, Guangdong, China.
| | - Weibin Cai
- Institute of Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
35
|
Berecz T, Yiu A, Vittay O, Orsolits B, Mioulane M, dos Remedios CG, Ketteler R, Merkely B, Apáti Á, Harding SE, Hellen N, Foldes G. Transcriptional co-activators YAP1-TAZ of Hippo signalling in doxorubicin-induced cardiomyopathy. ESC Heart Fail 2022; 9:224-235. [PMID: 34931757 PMCID: PMC8787991 DOI: 10.1002/ehf2.13756] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 11/02/2021] [Accepted: 11/24/2021] [Indexed: 11/07/2022] Open
Abstract
AIMS Hippo signalling is an evolutionarily conserved pathway that controls organ size by regulating apoptosis, cell proliferation, and stem cell self-renewal. Recently, the pathway has been shown to exert powerful growth regulatory activity in cardiomyocytes. However, the functional role of this stress-related and cell death-related pathway in the human heart and cardiomyocytes is not known. In this study, we investigated the role of the transcriptional co-activators of Hippo signalling, YAP and TAZ, in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in response to cardiotoxic agents and investigated the effects of modulating the pathway on cardiomyocyte function and survival. METHODS AND RESULTS RNA-sequencing analysis of human heart samples with doxorubicin-induced end-stage heart failure and healthy controls showed that YAP and ERBB2 (HER2) as upstream regulators of differentially expressed genes correlated with doxorubicin treatment. Thus, we tested the effects of doxorubicin on hiPSC-CMs in vitro. Using an automated high-content screen of 96 clinically relevant antineoplastic and cardiotherapeutic drugs, we showed that doxorubicin induced the highest activation of YAP/TAZ nuclear translocation in both hiPSC-CMs and control MCF7 breast cancer cells. The overexpression of YAP rescued doxorubicin-induced cell loss in hiPSC-CMs by inhibiting apoptosis and inducing proliferation. In contrast, silencing of YAP and TAZ by siRNAs resulted in elevated mitochondrial membrane potential loss in response to doxorubicin. hiPSC-CM calcium transients did not change in response to YAP/TAZ silencing. CONCLUSIONS Our results suggest that Hippo signalling is involved in clinical anthracycline-induced cardiomyopathy. Modelling with hiPSC-CMs in vitro showed similar responses to doxorubicin as adult cardiomyocytes and revealed a potential cardioprotective effect of YAP in doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Tünde Berecz
- Heart and Vascular CenterSemmelweis University68 Városmajor StreetBudapestH1122Hungary
- Institute of Enzymology, Research Centre for Natural SciencesEötvös Loránd Research NetworkBudapestHungary
| | - Angela Yiu
- Department of Surgery and CancerImperial College LondonLondonUK
| | - Orsolya Vittay
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Barbara Orsolits
- Heart and Vascular CenterSemmelweis University68 Városmajor StreetBudapestH1122Hungary
| | - Maxime Mioulane
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Cristobal G. dos Remedios
- Victor Chang Cardiac Research InstituteDarlinghurstNSWAustralia
- Bosch InstituteThe University of SydneySydneyNSWAustralia
| | - Robin Ketteler
- Laboratory for Molecular Cell BiologyUniversity College LondonLondonUK
| | - Bela Merkely
- Heart and Vascular CenterSemmelweis University68 Városmajor StreetBudapestH1122Hungary
| | - Ágota Apáti
- Institute of Enzymology, Research Centre for Natural SciencesEötvös Loránd Research NetworkBudapestHungary
| | - Sian E. Harding
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Nicola Hellen
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Gabor Foldes
- Heart and Vascular CenterSemmelweis University68 Városmajor StreetBudapestH1122Hungary
- National Heart and Lung InstituteImperial College LondonLondonUK
| |
Collapse
|
36
|
Okamura DM, Nguyen ED, Beier DR, Majesky MW. Wound healing and regeneration in spiny mice (Acomys cahirinus). Curr Top Dev Biol 2022; 148:139-164. [DOI: 10.1016/bs.ctdb.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
37
|
Uemasu H, Ikuta H, Igawa T, Suzuki M, Kyakuno M, Iwata Y, Tazawa I, Ogino H, Satoh Y, Takeuchi T, Namba N, Hayashi T. Cryo-injury procedure-induced cardiac regeneration shows unique gene expression profiles in the newt Pleurodeles waltl. Dev Dyn 2021; 251:864-876. [PMID: 34964213 DOI: 10.1002/dvdy.450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Cardiac regeneration in the adult mouse is not substantial. Some vertebrates, such as newts and zebrafish, regenerate the heart throughout their lives. To understand how regenerative abilities differ among animal species, comparative research has been conducted in animals like mouse, zebrafish, and newt. For those purposes, cryo-injury is suitable as an experimental model for the pathological condition of human myocardial infarction. In fact, cryo-injury procedures are common in mouse and zebrafish. RESULTS In the present study, we induced cryo-damage on the ventricle in Iberian ribbed newts using a liquid nitrogen-chilled probe. We observed that the injured area recovered within 8 weeks, with remodeling of scar tissue and proliferation of cardiomyocytes. We investigated the subsequent recovery of cryo-injured and amputated tissues by comparative analysis of the gene expression profiles following these two procedures. CONCLUSION Notably, we established a cryo-injury procedure for the newt and confirmed that regeneration of the cryo-damaged myocardial tissue is achieved by changes in gene expression that are milder than those observed in the amputation model. Our results suggest that the cryo-injury method is suitable for comparing the process of cardiac regeneration in the newt with that in other animal models. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Hitoshi Uemasu
- Division of Pediatrics and Perinatology, School of Medicine, Faculty of Medicine, Tottori University Yonago, Tottori, Japan
| | - Hiromi Ikuta
- Amphibian Research Center, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan.,Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Takeshi Igawa
- Amphibian Research Center, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan.,Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Makoto Suzuki
- Amphibian Research Center, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan.,Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Mitsuki Kyakuno
- Amphibian Research Center, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan.,Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Yui Iwata
- Amphibian Research Center, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan.,Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Ichiro Tazawa
- Amphibian Research Center, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan.,Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Hajime Ogino
- Amphibian Research Center, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan.,Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Yukio Satoh
- Department of Biomedical Sciences, School of Life Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| | - Takashi Takeuchi
- Department of Biomedical Sciences, School of Life Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| | - Noriyuki Namba
- Division of Pediatrics and Perinatology, School of Medicine, Faculty of Medicine, Tottori University Yonago, Tottori, Japan
| | - Toshinori Hayashi
- Amphibian Research Center, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan.,Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| |
Collapse
|
38
|
Quick RE, Buck LD, Parab S, Tolbert ZR, Matsuoka RL. Highly Efficient Synthetic CRISPR RNA/Cas9-Based Mutagenesis for Rapid Cardiovascular Phenotypic Screening in F0 Zebrafish. Front Cell Dev Biol 2021; 9:735598. [PMID: 34746131 PMCID: PMC8570140 DOI: 10.3389/fcell.2021.735598] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/01/2021] [Indexed: 12/13/2022] Open
Abstract
The zebrafish is a valuable vertebrate model to study cardiovascular formation and function due to the facile visualization and rapid development of the circulatory system in its externally growing embryos. Despite having distinct advantages, zebrafish have paralogs of many important genes, making reverse genetics approaches inefficient since generating animals bearing multiple gene mutations requires substantial efforts. Here, we present a simple and robust synthetic CRISPR RNA/Cas9-based mutagenesis approach for generating biallelic F0 zebrafish knockouts. Using a dual-guide synthetic CRISPR RNA/Cas9 ribonucleoprotein (dgRNP) system, we compared the efficiency of biallelic gene disruptions following the injections of one, two, and three dgRNPs per gene into the cytoplasm or yolk. We show that simultaneous cytoplasmic injections of three distinct dgRNPs per gene into one-cell stage embryos resulted in the most efficient and consistent biallelic gene disruptions. Importantly, this triple dgRNP approach enables efficient inactivation of cell autonomous and cell non-autonomous gene function, likely due to the low mosaicism of biallelic disruptions. In support of this finding, we provide evidence that the F0 animals generated by this method fully phenocopied the endothelial and peri-vascular defects observed in corresponding stable mutant homozygotes. Moreover, this approach faithfully recapitulated the trunk vessel phenotypes resulting from the genetic interaction between two vegfr2 zebrafish paralogs. Mechanistically, investigation of genome editing and mRNA decay indicates that the combined mutagenic actions of three dgRNPs per gene lead to an increased probability of frameshift mutations, enabling efficient biallelic gene disruptions. Therefore, our approach offers a highly robust genetic platform to quickly assess novel and redundant gene function in F0 zebrafish.
Collapse
Affiliation(s)
- Rachael E Quick
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Luke D Buck
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Sweta Parab
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Zane R Tolbert
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Ryota L Matsuoka
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
39
|
Bühler A, Gahr BM, Park DD, Bertozzi A, Boos A, Dalvoy M, Pott A, Oswald F, Kovall RA, Kühn B, Weidinger G, Rottbauer W, Just S. Histone deacetylase 1 controls cardiomyocyte proliferation during embryonic heart development and cardiac regeneration in zebrafish. PLoS Genet 2021; 17:e1009890. [PMID: 34723970 PMCID: PMC8584950 DOI: 10.1371/journal.pgen.1009890] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 11/11/2021] [Accepted: 10/18/2021] [Indexed: 12/20/2022] Open
Abstract
In contrast to mammals, the zebrafish maintains its cardiomyocyte proliferation capacity throughout adulthood. However, neither the molecular mechanisms that orchestrate the proliferation of cardiomyocytes during developmental heart growth nor in the context of regeneration in the adult are sufficiently defined yet. We identified in a forward genetic N-ethyl-N-nitrosourea (ENU) mutagenesis screen the recessive, embryonic-lethal zebrafish mutant baldrian (bal), which shows severely impaired developmental heart growth due to diminished cardiomyocyte proliferation. By positional cloning, we identified a missense mutation in the zebrafish histone deacetylase 1 (hdac1) gene leading to severe protein instability and the loss of Hdac1 function in vivo. Hdac1 inhibition significantly reduces cardiomyocyte proliferation, indicating a role of Hdac1 during developmental heart growth in zebrafish. To evaluate whether developmental and regenerative Hdac1-associated mechanisms of cardiomyocyte proliferation are conserved, we analyzed regenerative cardiomyocyte proliferation after Hdac1 inhibition at the wound border zone in cryoinjured adult zebrafish hearts and we found that Hdac1 is also essential to orchestrate regenerative cardiomyocyte proliferation in the adult vertebrate heart. In summary, our findings suggest an important and conserved role of Histone deacetylase 1 (Hdac1) in developmental and adult regenerative cardiomyocyte proliferation in the vertebrate heart. Heart disease is one of the most common causes of death in all developed countries. While zebrafish cardiomyocytes are able to proliferate throughout adulthood, mammalian cardiomyocytes lose this ability during early development, and therefore are not capable to replace and renew cardiomyocytes after injury. The underlying mechanisms of cardiomyocyte proliferation are still not completely resolved. Understanding how zebrafish cardiomyocytes preserve their proliferating state, would be a valuable information to foster cardiac regeneration, e.g. after myocardial infarction in patients. Knowledge of the signaling pathways that need to be activated, or deactivated in order to induce cardiomyocyte proliferation after acute or chronic injury will pave the way for the development of genetic and/or pharmacological treatment options. In an ENU-mutagenesis screen, we identified the zebrafish mutant baldrian, which shows reduced embryonic cardiomyocyte proliferation. As genetic cause of the observed phenotype, we identified a missense mutation in the hdac1 gene. By treatment of heart-injured adult fish with the HDAC1 inhibitor Mocetinostat, we were able to show a reduced rate of cardiomyocyte proliferation also in the adult zebrafish heart in vivo, suggesting a role of Hdac1 in embryonic heart growth and adult regenerative cardiomyocyte proliferation in zebrafish.
Collapse
Affiliation(s)
- Anja Bühler
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Bernd M Gahr
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Deung-Dae Park
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Alberto Bertozzi
- Institute of Biochemistry and Molecular Biology, University of Ulm, Ulm, Germany
| | - Alena Boos
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Mohankrishna Dalvoy
- Institute of Biochemistry and Molecular Biology, University of Ulm, Ulm, Germany
| | - Alexander Pott
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany.,Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Franz Oswald
- Department of Internal Medicine I, University of Ulm, Ulm, Germany
| | - Rhett A Kovall
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Bernhard Kühn
- Department of Pediatrics, University of Pittsburgh, and Richard King Mellon Institute for Pediatric Research and Division of Pediatric Cardiology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, United States of America
| | - Gilbert Weidinger
- Institute of Biochemistry and Molecular Biology, University of Ulm, Ulm, Germany
| | | | - Steffen Just
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| |
Collapse
|
40
|
Pant T, Juric M, Bosnjak ZJ, Dhanasekaran A. Recent Insight on the Non-coding RNAs in Mesenchymal Stem Cell-Derived Exosomes: Regulatory and Therapeutic Role in Regenerative Medicine and Tissue Engineering. Front Cardiovasc Med 2021; 8:737512. [PMID: 34660740 PMCID: PMC8517144 DOI: 10.3389/fcvm.2021.737512] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/06/2021] [Indexed: 12/13/2022] Open
Abstract
Advances in the field of regenerative medicine and tissue engineering over the past few decades have paved the path for cell-free therapy. Numerous stem cell types, including mesenchymal stem cells (MSCs), have been reported to impart therapeutic effects via paracrine secretion of exosomes. The underlying factors and the associated mechanisms contributing to these MSC-derived exosomes' protective effects are, however, poorly understood, limiting their application in the clinic. The exosomes exhibit a diversified repertoire of functional non-coding RNAs (ncRNAs) and have the potential to transfer these biologically active transcripts to the recipient cells, where they are found to modulate a diverse array of functions. Altered expression of the ncRNAs in the exosomes has been linked with the regenerative potential and development of various diseases, including cardiac, neurological, skeletal, and cancer. Also, modulating the expression of ncRNAs in these exosomes has been found to improve their therapeutic impact. Moreover, many of these ncRNAs are expressed explicitly in the MSC-derived exosomes, making them ideal candidates for regenerative medicine, including tissue engineering research. In this review, we detail the recent advances in regenerative medicine and summarize the evidence supporting the altered expression of the ncRNA repertoire specific to MSCs under different degenerative diseases. We also discuss the therapeutic role of these ncRNA for the prevention of these various degenerative diseases and their future in translational medicine.
Collapse
Affiliation(s)
- Tarun Pant
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Matea Juric
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Zeljko J. Bosnjak
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | | |
Collapse
|
41
|
Ivankovic I, Déan-Ben XL, Haas H, Kimm MA, Wildgruber M, Razansky D. Volumetric Optoacoustic Tomography Differentiates Myocardial Remodelling. Mol Imaging Biol 2021; 22:1235-1243. [PMID: 32394284 DOI: 10.1007/s11307-020-01498-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE Myocardial healing following myocardial infarction (MI) is a complex process that is yet to be fully understood. Clinical attempts in regeneration of the injured myocardium using cardiac stem cells faced major challenges, calling for a better understanding of the processes involved at a more basic level in order to foster translation. PROCEDURES We examined the feasibility of volumetric optoacoustic tomography (VOT) in studying healing of the myocardium in different models of MI, including permanent occlusion (PO) of the left coronary artery, temporary occlusion (ischemia-reperfusion-I/R) and infarcted c-kit mutants, a genetic mouse model with impaired cardiac healing. Murine hearts were imaged at 100 Hz frame rate using 800 nm excitation wavelength, corresponding to the peak absorption of indocyanine green (ICG) in plasma and the isosbestic point of haemoglobin. RESULTS The non-invasive real-time volumetric imaging capabilities of VOT have allowed the detection of significant variations in the pulmonary transit time (PTT), a parameter affected by MI, across different murine models. Upon intravenous injection of ICG, we were able to track alterations in cardiac perfusion in I/R models, which were absent in wild-type (wt) PO or kitW/kitW-v PO mice. The wt-PO and I/R models further exhibited irregularities in their cardiac cycles. CONCLUSIONS Clear differences in the PTT, ICG perfusion and cardiac cycle patterns were identified between the different models and days post MI. Overall, the results highlight the unique capacity of VOT for multi-parametric characterization of morphological and functional changes in murine models of MI.
Collapse
Affiliation(s)
- Ivana Ivankovic
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering and Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Xosé Luís Déan-Ben
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering and Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Helena Haas
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar Technical University of Munich, Munich, Germany
| | - Melanie A Kimm
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar Technical University of Munich, Munich, Germany
| | - Moritz Wildgruber
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar Technical University of Munich, Munich, Germany
- Translational Research Imaging Center, Department of Clinical Radiology, Universitätsklinikum Münster, Munster, Germany
| | - Daniel Razansky
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
- Institute for Biomedical Engineering and Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
42
|
Shimizu Y, Kawasaki T. Differential Regenerative Capacity of the Optic Tectum of Adult Medaka and Zebrafish. Front Cell Dev Biol 2021; 9:686755. [PMID: 34268310 PMCID: PMC8276636 DOI: 10.3389/fcell.2021.686755] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/01/2021] [Indexed: 12/03/2022] Open
Abstract
Zebrafish have superior regenerative capacity in the central nervous system (CNS) compared to mammals. In contrast, medaka were shown to have low regenerative capacity in the adult heart and larval retina, despite the well-documented high tissue regenerative ability of teleosts. Nevertheless, medaka and zebrafish share similar brain structures and biological features to those of mammals. Hence, this study aimed to compare the neural stem cell (NSC) responses and regenerative capacity in the optic tectum of adult medaka and zebrafish after stab wound injury. Limited neuronal differentiation was observed in the injured medaka, though the proliferation of radial glia (RG) was induced in response to tectum injury. Moreover, the expression of the pro-regenerative transcriptional factors ascl1a and oct4 was not enhanced in the injured medaka, unlike in zebrafish, whereas expression of sox2 and stat3 was upregulated in both fish models. Of note, glial scar-like structures composed of GFAP+ radial fibers were observed in the injured area of medaka at 14 days post injury (dpi). Altogether, these findings suggest that the adult medaka brain has low regenerative capacity with limited neuronal generation and scar formation. Hence, medaka represent an attractive model for investigating and evaluating critical factors for brain regeneration.
Collapse
Affiliation(s)
- Yuki Shimizu
- Functional Biomolecular Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Osaka, Japan
- DBT-AIST International Laboratory for Advanced Biomedicine, National Institute of Advanced Industrial Science and Technology, Osaka, Japan
| | - Takashi Kawasaki
- Functional Biomolecular Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Osaka, Japan
| |
Collapse
|
43
|
Khosravi F, Ahmadvand N, Bellusci S, Sauer H. The Multifunctional Contribution of FGF Signaling to Cardiac Development, Homeostasis, Disease and Repair. Front Cell Dev Biol 2021; 9:672935. [PMID: 34095143 PMCID: PMC8169986 DOI: 10.3389/fcell.2021.672935] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022] Open
Abstract
The current focus on cardiovascular research reflects society’s concerns regarding the alarming incidence of cardiac-related diseases and mortality in the industrialized world and, notably, an urgent need to combat them by more efficient therapies. To pursue these therapeutic approaches, a comprehensive understanding of the mechanism of action for multifunctional fibroblast growth factor (FGF) signaling in the biology of the heart is a matter of high importance. The roles of FGFs in heart development range from outflow tract formation to the proliferation of cardiomyocytes and the formation of heart chambers. In the context of cardiac regeneration, FGFs 1, 2, 9, 16, 19, and 21 mediate adaptive responses including restoration of cardiac contracting rate after myocardial infarction and reduction of myocardial infarct size. However, cardiac complications in human diseases are correlated with pathogenic effects of FGF ligands and/or FGF signaling impairment. FGFs 2 and 23 are involved in maladaptive responses such as cardiac hypertrophic, fibrotic responses and heart failure. Among FGFs with known causative (FGFs 2, 21, and 23) or protective (FGFs 2, 15/19, 16, and 21) roles in cardiac diseases, FGFs 15/19, 21, and 23 display diagnostic potential. The effective role of FGFs on the induction of progenitor stem cells to cardiac cells during development has been employed to boost the limited capacity of postnatal cardiac repair. To renew or replenish damaged cardiomyocytes, FGFs 1, 2, 10, and 16 were tested in (induced-) pluripotent stem cell-based approaches and for stimulation of cell cycle re-entry in adult cardiomyocytes. This review will shed light on the wide range of beneficiary and detrimental actions mediated by FGF ligands and their receptors in the heart, which may open new therapeutic avenues for ameliorating cardiac complications.
Collapse
Affiliation(s)
- Farhad Khosravi
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Negah Ahmadvand
- Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| | - Saverio Bellusci
- Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| | - Heinrich Sauer
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
44
|
Electroactive Polymeric Composites to Mimic the Electromechanical Properties of Myocardium in Cardiac Tissue Repair. Gels 2021; 7:gels7020053. [PMID: 34062741 PMCID: PMC8162334 DOI: 10.3390/gels7020053] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
Due to the limited regenerative capabilities of cardiomyocytes, incidents of myocardial infarction can cause permanent damage to native myocardium through the formation of acellular, non-conductive scar tissue during wound repair. The generation of scar tissue in the myocardium compromises the biomechanical and electrical properties of the heart which can lead to further cardiac problems including heart failure. Currently, patients suffering from cardiac failure due to scarring undergo transplantation but limited donor availability and complications (i.e., rejection or infectious pathogens) exclude many individuals from successful transplant. Polymeric tissue engineering scaffolds provide an alternative approach to restore normal myocardium structure and function after damage by acting as a provisional matrix to support cell attachment, infiltration and stem cell delivery. However, issues associated with mechanical property mismatch and the limited electrical conductivity of these constructs when compared to native myocardium reduces their clinical applicability. Therefore, composite polymeric scaffolds with conductive reinforcement components (i.e., metal, carbon, or conductive polymers) provide tunable mechanical and electroactive properties to mimic the structure and function of natural myocardium in force transmission and electrical stimulation. This review summarizes recent advancements in the design, synthesis, and implementation of electroactive polymeric composites to better match the biomechanical and electrical properties of myocardial tissue.
Collapse
|
45
|
Peralta M, Ortiz Lopez L, Jerabkova K, Lucchesi T, Vitre B, Han D, Guillemot L, Dingare C, Sumara I, Mercader N, Lecaudey V, Delaval B, Meilhac SM, Vermot J. Intraflagellar Transport Complex B Proteins Regulate the Hippo Effector Yap1 during Cardiogenesis. Cell Rep 2021; 32:107932. [PMID: 32698004 DOI: 10.1016/j.celrep.2020.107932] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 04/30/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023] Open
Abstract
Cilia and the intraflagellar transport (IFT) proteins involved in ciliogenesis are associated with congenital heart diseases (CHDs). However, the molecular links between cilia, IFT proteins, and cardiogenesis are yet to be established. Using a combination of biochemistry, genetics, and live-imaging methods, we show that IFT complex B proteins (Ift88, Ift54, and Ift20) modulate the Hippo pathway effector YAP1 in zebrafish and mouse. We demonstrate that this interaction is key to restrict the formation of the proepicardium and the myocardium. In cellulo experiments suggest that IFT88 and IFT20 interact with YAP1 in the cytoplasm and functionally modulate its activity, identifying a molecular link between cilia-related proteins and the Hippo pathway. Taken together, our results highlight a noncanonical role for IFT complex B proteins during cardiogenesis and shed light on a mechanism of action for ciliary proteins in YAP1 regulation.
Collapse
Affiliation(s)
- Marina Peralta
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France; Université de Strasbourg, Illkirch, France
| | - Laia Ortiz Lopez
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France; Université de Strasbourg, Illkirch, France
| | - Katerina Jerabkova
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France; Université de Strasbourg, Illkirch, France
| | - Tommaso Lucchesi
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, Paris, France; INSERM UMR1163, Université Paris Descartes, Paris, France; Sorbonne Université, Collège Doctoral, F-75005, Paris, France
| | - Benjamin Vitre
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), CNRS, Université de Montpellier, Montpellier, France
| | - Dong Han
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, Paris, France; INSERM UMR1163, Université Paris Descartes, Paris, France
| | - Laurent Guillemot
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, Paris, France; INSERM UMR1163, Université Paris Descartes, Paris, France
| | - Chaitanya Dingare
- Institute for Cell Biology and Neurosciences, Goethe University of Frankfurt, Frankfurt, Germany
| | - Izabela Sumara
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France; Université de Strasbourg, Illkirch, France
| | - Nadia Mercader
- Institute of Anatomy, University of Bern, Bern, Switzerland; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Virginie Lecaudey
- Institute for Cell Biology and Neurosciences, Goethe University of Frankfurt, Frankfurt, Germany
| | - Benedicte Delaval
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), CNRS, Université de Montpellier, Montpellier, France
| | - Sigolène M Meilhac
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, Paris, France; INSERM UMR1163, Université Paris Descartes, Paris, France
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France; Université de Strasbourg, Illkirch, France; Sorbonne Université, Collège Doctoral, F-75005, Paris, France; Department of Bioengineering, Imperial College London, London, UK.
| |
Collapse
|
46
|
Deyell M, Garris CS, Laughney AM. Cancer metastasis as a non-healing wound. Br J Cancer 2021; 124:1491-1502. [PMID: 33731858 PMCID: PMC8076293 DOI: 10.1038/s41416-021-01309-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 01/25/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023] Open
Abstract
Most cancer deaths are caused by metastasis: recurrence of disease by disseminated tumour cells at sites distant from the primary tumour. Large numbers of disseminated tumour cells are released from the primary tumour, even during the early stages of tumour growth. However, only a minority survive as potential seeds for future metastatic outgrowths. These cells must adapt to a relatively inhospitable microenvironment, evade immune surveillance and progress from the micro- to macro-metastatic stage to generate a secondary tumour. A pervasive driver of this transition is chronic inflammatory signalling emanating from tumour cells themselves. These signals can promote migration and engagement of stem and progenitor cell function, events that are also central to a wound healing response. In this review, we revisit the concept of cancer as a non-healing wound, first introduced by Virchow in the 19th century, with a new tumour cell-intrinsic perspective on inflammation and focus on metastasis. Cellular responses to inflammation in both wound healing and metastasis are tightly regulated by crosstalk with the surrounding microenvironment. Targeting or restoring canonical responses to inflammation could represent a novel strategy to prevent the lethal spread of cancer.
Collapse
Affiliation(s)
- Matthew Deyell
- grid.5386.8000000041936877XInstitute for Computational Biomedicine, Weill Cornell Medicine, New York, NY USA ,grid.5386.8000000041936877XDepartment of Physiology and Biophysics, Weill Cornell Medicine, New York, NY USA ,grid.5386.8000000041936877XSandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY USA ,grid.4444.00000 0001 2112 9282Chimie Biologie et Innovation, ESPCI Paris, Université PSL, CNRS, Paris, France
| | | | - Ashley M. Laughney
- grid.5386.8000000041936877XInstitute for Computational Biomedicine, Weill Cornell Medicine, New York, NY USA ,grid.5386.8000000041936877XDepartment of Physiology and Biophysics, Weill Cornell Medicine, New York, NY USA ,grid.5386.8000000041936877XSandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY USA
| |
Collapse
|
47
|
Kit-Anan W, Mazo MM, Wang BX, Leonardo V, Pence IJ, Gopal S, Gelmi A, Nagelkerke A, Becce M, Chiappini C, Harding SE, Terracciano CM, Stevens MM. Multiplexing physical stimulation on single human induced pluripotent stem cell-derived cardiomyocytes for phenotype modulation. Biofabrication 2021; 13:025004. [PMID: 33710972 PMCID: PMC7610872 DOI: 10.1088/1758-5090/abce0a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/11/2020] [Accepted: 11/25/2020] [Indexed: 12/02/2022]
Abstract
Traditional in vitro bioengineering approaches whereby only individual biophysical cues are manipulated at any one time are highly inefficient, falling short when recapitulating the complexity of the cardiac environment. Multiple biophysical cues are present in the native myocardial niche and are essential during development, as well as in maintenance of adult cardiomyocyte (CM) phenotype in both health and disease. This study establishes a novel biofabrication workflow to study and manipulate hiPSC-CMs and to understand how these cells respond to a multiplexed biophysical environment, namely 3D shape and substrate stiffness, at a single cell level. Silicon masters were fabricated and developed to generate inverse patterns of the desired 3D shapes in bas relief, which then were used to mold the designed microwell arrays into a hydrogel. Polyacrylamide (PAAm) was modified with the incorporation of acrylic acid to provide a carboxylic group conjugation site for adhesion motifs, without compromising capacity to modulate stiffness. In this manner, two individual parameters can be finely tuned independently within the hydrogel: the shape of the 3D microwell and its stiffness. The design allows the platform to isolate single hiPSC-CMs to study solely biophysical cues in the absence of cell-cell physical interaction. Under physiologic-like physical conditions (3D shape resembling that of adult CM and 9.83 kPa substrate stiffness that mimics muscle stiffness), isolated single hiPSC-CMs exhibit increased Cx-43 density, cell membrane stiffness and calcium transient amplitude; co-expression of the subpopulation-related MYL2-MYL7 proteins; and higher anisotropism than cells in pathologic-like conditions (flat surface and 112 kPa substrate stiffness). This demonstrates that supplying a physiologic or pathologic microenvironment to an isolated single hiPSC-CM in the absence of any physical cell-to-cell communication in this biofabricated platform leads to a significantly different set of cellular features, thus presenting a differential phenotype. Importantly, this demonstrates the high plasticity of hiPSC-CMs even in isolation. The ability of multiple biophysical cues to significantly influence isolated single hiPSC-CM phenotype and functionality highlights the importance of fine-tuning such cues for specific applications. This has the potential to produce more fit-for-purpose hiPSC-CMs. Further understanding of human cardiac development is enabled by the robust, versatile and reproducible biofabrication techniques applied here. We envision that this system could be easily applied to other tissues and cell types where the influence of cellular shape and stiffness of the surrounding environment is hypothesized to play an important role in physiology.
Collapse
Affiliation(s)
- Worrapong Kit-Anan
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Manuel M Mazo
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Brian X Wang
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Vincent Leonardo
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Isaac J Pence
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Sahana Gopal
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Amy Gelmi
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
- Current Address: Applied Chemistry and Environmental Science, School of Science, RMIT University, Melbourne, VIC 3001, Australia
| | - Anika Nagelkerke
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Michele Becce
- Department of Materials, Imperial College London, London, United Kingdom
| | - Ciro Chiappini
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Sian E Harding
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Cesare M Terracciano
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Molly M Stevens
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| |
Collapse
|
48
|
Quadri F, Soman SS, Vijayavenkataraman S. Progress in cardiovascular bioprinting. Artif Organs 2021; 45:652-664. [PMID: 33432583 DOI: 10.1111/aor.13913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/13/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease has been the leading cause of death globally for the past 15 years. Following a major cardiac disease episode, the ideal treatment would be the replacement of the damaged tissue, due to the limited regenerative capacity of cardiac tissues. However, we suffer from a chronic organ donor shortage which causes approximately 20 people to die each day waiting to receive an organ. Bioprinting of tissues and organs can potentially alleviate this burden by fabricating low cost tissue and organ replacements for cardiac patients. Clinical adoption of bioprinting in cardiovascular medicine is currently limited by the lack of systematic demonstration of its effectiveness, high costs, and the complexity of the workflow. Here, we give a concise review of progress in cardiovascular bioprinting and its components. We further discuss the challenges and future prospects of cardiovascular bioprinting in clinical applications.
Collapse
Affiliation(s)
- Faisal Quadri
- Division of Science, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Soja Saghar Soman
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Sanjairaj Vijayavenkataraman
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, UAE.,Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, USA
| |
Collapse
|
49
|
Peng H, Chelvarajan L, Donahue R, Gottipati A, Cahall CF, Davis KA, Tripathi H, Al-Darraji A, Elsawalhy E, Dobrozsi N, Srinivasan A, Levitan BM, Kong R, Gao E, Abdel-Latif A, Berron BJ. Polymer Cell Surface Coating Enhances Mesenchymal Stem Cell Retention and Cardiac Protection. ACS APPLIED BIO MATERIALS 2021; 4:1655-1667. [PMID: 35014513 DOI: 10.1021/acsabm.0c01473] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Mesenchymal stem cell (MSC) therapy has been widely tested in clinical trials to promote healing post-myocardial infarction. However, low cell retention and the need for a large donor cell number in human studies remain a key challenge for clinical translation. Natural biomaterials such as gelatin are ideally suited as scaffolds to deliver and enhance cell engraftment after transplantation. A potential drawback of MSC encapsulation in the hydrogel is that the bulky matrix may limit their biological function and interaction with the surrounding tissue microenvironment that conveys important injury signals. To overcome this limitation, we adopted a gelatin methacrylate (gelMA) cell-coating technique that photocross-links gelatin on the individual cell surface at the nanoscale. The present study investigated the cardiac protection of gelMA coated, hypoxia preconditioned MSCs (gelMA-MSCs) in a murine myocardial infarction (MI) model. We demonstrate that the direct injection of gelMA-MSC results in significantly higher myocardial engraftment 7 days after MI compared to uncoated MSCs. GelMA-MSC further amplified MSC benefits resulting in enhanced cardioprotection as measured by cardiac function, scar size, and angiogenesis. Improved MSC cardiac retention also led to a greater cardiac immunomodulatory function after injury. Taken together, this study demonstrated the efficacy of gelMA-MSCs in treating cardiac injury with a promising potential to reduce the need for donor MSCs through enhanced myocardial engraftment.
Collapse
Affiliation(s)
- Hsuan Peng
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States
| | - Lakshman Chelvarajan
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States
| | - Renee Donahue
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States
| | - Anuhya Gottipati
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Calvin F Cahall
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Kara A Davis
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Himi Tripathi
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States
| | - Ahmed Al-Darraji
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States
| | - Eman Elsawalhy
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States
| | - Nicholas Dobrozsi
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States
| | - Amrita Srinivasan
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States
| | - Bryana M Levitan
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States.,Department of Physiology, University of Kentucky, Lexington, Kentucky 40508, United States
| | - Raymond Kong
- MilliporeSigma, Seattle, Washington 98119, United States
| | - Erhe Gao
- The Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, United States
| | - Ahmed Abdel-Latif
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States
| | - Brad J Berron
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky 40506, United States
| |
Collapse
|
50
|
Knecht RS, Bucher CH, Van Linthout S, Tschöpe C, Schmidt-Bleek K, Duda GN. Mechanobiological Principles Influence the Immune Response in Regeneration: Implications for Bone Healing. Front Bioeng Biotechnol 2021; 9:614508. [PMID: 33644014 PMCID: PMC7907627 DOI: 10.3389/fbioe.2021.614508] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/22/2021] [Indexed: 12/13/2022] Open
Abstract
A misdirected or imbalanced local immune composition is often one of the reasons for unsuccessful regeneration resulting in scarring or fibrosis. Successful healing requires a balanced initiation and a timely down-regulation of the inflammation for the re-establishment of a biologically and mechanically homeostasis. While biomaterial-based approaches to control local immune responses are emerging as potential new treatment options, the extent to which biophysical material properties themselves play a role in modulating a local immune niche response has so far been considered only occasionally. The communication loop between extracellular matrix, non-hematopoietic cells, and immune cells seems to be specifically sensitive to mechanical cues and appears to play a role in the initiation and promotion of a local inflammatory setting. In this review, we focus on the crosstalk between ECM and its mechanical triggers and how they impact immune cells and non-hematopoietic cells and their crosstalk during tissue regeneration. We realized that especially mechanosensitive receptors such as TRPV4 and PIEZO1 and the mechanosensitive transcription factor YAP/TAZ are essential to regeneration in various organ settings. This indicates novel opportunities for therapeutic approaches to improve tissue regeneration, based on the immune-mechanical principles found in bone but also lung, heart, and skin.
Collapse
Affiliation(s)
- Raphael S Knecht
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christian H Bucher
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Carsten Tschöpe
- Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany.,Department of Cardiology, Charite'-Universitätsmedizin Berlin, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Georg N Duda
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|