1
|
Masciale V, Banchelli F, Grisendi G, Samarelli AV, Raineri G, Rossi T, Zanoni M, Cortesi M, Bandini S, Ulivi P, Martinelli G, Stella F, Dominici M, Aramini B. The molecular features of lung cancer stem cells in dedifferentiation process-driven epigenetic alterations. J Biol Chem 2024; 300:107994. [PMID: 39547513 PMCID: PMC11714729 DOI: 10.1016/j.jbc.2024.107994] [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: 03/23/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Cancer stem cells (CSCs) may be dedifferentiated somatic cells following oncogenic processes, representing a subpopulation of cells able to promote tumor growth with their capacities for proliferation and self-renewal, inducing lineage heterogeneity, which may be a main cause of resistance to therapies. It has been shown that the "less differentiated process" may have an impact on tumor plasticity, particularly when non-CSCs may dedifferentiate and become CSC-like. Bidirectional interconversion between CSCs and non-CSCs has been reported in other solid tumors, where the inflammatory stroma promotes cell reprogramming by enhancing Wnt signaling through nuclear factor kappa B activation in association with intracellular signaling, which may induce cells' pluripotency, the oncogenic transformation can be considered another important aspect in the acquisition of "new" development programs with oncogenic features. During cell reprogramming, mutations represent an initial step toward dedifferentiation, in which tumor cells switch from a partially or terminally differentiated stage to a less differentiated stage that is mainly manifested by re-entry into the cell cycle, acquisition of a stem cell-like phenotype, and expression of stem cell markers. This phenomenon typically shows up as a change in the form, function, and pattern of gene and protein expression, and more specifically, in CSCs. This review would highlight the main epigenetic alterations, major signaling pathways and driver mutations in which CSCs, in tumors and specifically, in lung cancer, could be involved, acting as key elements in the differentiation/dedifferentiation process. This would highlight the main molecular mechanisms which need to be considered for more tailored therapies.
Collapse
Affiliation(s)
- Valentina Masciale
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Federico Banchelli
- Department of Statistical Sciences "Paolo Fortunati", Alma Mater Studiorum- University of Bologna, Bologna, Italy
| | - Giulia Grisendi
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Anna Valeria Samarelli
- Laboratory of and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Giulia Raineri
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Tania Rossi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Michele Zanoni
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Michela Cortesi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sara Bandini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Paola Ulivi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giovanni Martinelli
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Franco Stella
- Thoracic Surgery Unit, Department of Medical and Surgical Sciences-DIMEC of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni-L. Pierantoni Hospital, Forlì, Italy
| | - Massimo Dominici
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy; Division of Oncology, University Hospital of Modena and Reggio Emilia, University of Modena and Reggio Emilia, Modena, Italy
| | - Beatrice Aramini
- Thoracic Surgery Unit, Department of Medical and Surgical Sciences-DIMEC of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni-L. Pierantoni Hospital, Forlì, Italy.
| |
Collapse
|
2
|
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
|
3
|
Sebastian WA, Inoue M, Shimizu N, Sato R, Oguri S, Itonaga T, Kishimoto S, Shiraishi H, Hanada T, Ihara K. Cardiac manifestations of human ACTA2 variants recapitulated in a zebrafish model. J Hum Genet 2024; 69:133-138. [PMID: 38316882 DOI: 10.1038/s10038-024-01221-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/31/2023] [Accepted: 01/19/2024] [Indexed: 02/07/2024]
Abstract
The ACTA2 gene encodes actin α2, a major smooth muscle protein in vascular smooth muscle cells. Missense variants in the ACTA2 gene can cause inherited thoracic aortic diseases with characteristic symptoms, such as dysfunction of smooth muscle cells in the lungs, brain vessels, intestines, pupils, bladder, or heart. We identified a heterozygous missense variant of Gly148Arg (G148R) in a patient with a thoracic aortic aneurysm, dissection, and left ventricular non-compaction. We used zebrafish as an in vivo model to investigate whether or not the variants might cause functional or histopathological abnormalities in the heart. Following the fertilization of one-cell stage embryos, we injected in vitro synthesized ACTA2 mRNA of wild-type, novel variant G148R, or the previously known pathogenic variant Arg179His (R179H). The embryos were maintained and raised for 72 h post-fertilization for a heart analysis. Shortening fractions of heart were significantly reduced in both pathogenic variants. A histopathological evaluation showed that the myocardial wall of ACTA2 pathogenic variants was thinner than that of the wild type, and the total cell number within the myocardium was markedly decreased in all zebrafish with pathogenic variants mRNAs. Proliferating cell numbers were also significantly decreased in the endothelial and myocardial regions of zebrafish with ACTA2 variants compared to the wild type. These results demonstrate the effects of ACTA2 G148R and R179H on the development of left ventricle non-compaction and cardiac morphological abnormalities. Our study highlights the previously unknown significance of the ACTA2 gene in several aspects of cardiovascular development.
Collapse
Affiliation(s)
| | - Masanori Inoue
- Department of Pediatrics, Oita University, Faculty of Medicine, Oita, Japan
| | - Nobuyuki Shimizu
- Department of Cell Biology, Oita University, Faculty of Medicine, Oita, Japan
| | - Ryosuke Sato
- Department of Pediatrics, Oita University, Faculty of Medicine, Oita, Japan
| | - Saori Oguri
- Department of Pediatrics, Oita University, Faculty of Medicine, Oita, Japan
| | - Tomoyo Itonaga
- Department of Pediatrics, Oita University, Faculty of Medicine, Oita, Japan
| | - Shintaro Kishimoto
- Department of Pediatrics, Oita University, Faculty of Medicine, Oita, Japan
| | - Hiroshi Shiraishi
- Department of Cell Biology, Oita University, Faculty of Medicine, Oita, Japan
| | - Toshikatsu Hanada
- Department of Cell Biology, Oita University, Faculty of Medicine, Oita, Japan.
| | - Kenji Ihara
- Department of Pediatrics, Oita University, Faculty of Medicine, Oita, Japan.
| |
Collapse
|
4
|
Huo Y, Wang W, Zhang J, Xu D, Bai F, Gui Y. Maternal androgen excess inhibits fetal cardiomyocytes proliferation through RB-mediated cell cycle arrest and induces cardiac hypertrophy in adulthood. J Endocrinol Invest 2024; 47:603-617. [PMID: 37642904 PMCID: PMC10904501 DOI: 10.1007/s40618-023-02178-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023]
Abstract
PURPOSE Maternal hyperandrogenism during pregnancy is associated with adverse gestational outcomes and chronic non-communicable diseases in offspring. However, few studies are reported to demonstrate the association between maternal androgen excess and cardiac health in offspring. This study aimed to explore the relation between androgen exposure in utero and cardiac health of offspring in fetal and adult period. Its underlying mechanism is also illustrated in this research. METHODS Pregnant mice were injected with dihydrotestosterone (DHT) from gestational day (GD) 16.5 to GD18.5. On GD18.5, fetal heart tissue was collected for metabolite and morphological analysis. The hearts from adult offspring were also collected for morphological and qPCR analysis. H9c2 cells were treated with 75 μM androsterone. Immunofluorescence, flow cytometry, qPCR, and western blot were performed to observe cell proliferation and explore the underlying mechanism. RESULTS Intrauterine exposure to excessive androgen led to thinner ventricular wall, decreased number of cardiomyocytes in fetal offspring and caused cardiac hypertrophy, compromised cardiac function in adult offspring. The analysis of steroid hormone metabolites in fetal heart tissue by ultra performance liquid chromatography and tandem mass spectrometry showed that the content of androgen metabolite androsterone was significantly increased. Mechanistically, H9c2 cells treated with androsterone led to a significant decrease in phosphorylated retinoblastoma protein (pRB) and cell cycle-related protein including cyclin-dependent kinase 2 (CDK2), cyclin-dependent kinase 4 (CDK4), and cyclin D1 (CCND1) in cardiomyocytes. This resulted in cell cycle arrest at G1-S phase, which in turn inhibited cardiomyocyte proliferation. CONCLUSION Taken together, our results indicate that in utero exposure to DHT, its metabolite androsterone could directly decrease cardiomyocytes proliferation through cell cycle arrest, which has a life-long-lasting effect on cardiac health. Our study highlights the importance of monitoring sex hormones in women during pregnancy and the follow-up of cardiac function in offspring with high risk of intrauterine androgen exposure.
Collapse
Affiliation(s)
- Y Huo
- National Children's Medical Center, Children's Hospital of Fudan University, Fudan University, Shanghai, 201102, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, 399 Wanyuan Road, Minhang, Shanghai, 201102, China
| | - W Wang
- Guangzhou Center for Disease Control and Prevention, Guangzhou, 510080, China
| | - J Zhang
- National Children's Medical Center, Children's Hospital of Fudan University, Fudan University, Shanghai, 201102, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, 399 Wanyuan Road, Minhang, Shanghai, 201102, China
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - D Xu
- National Children's Medical Center, Children's Hospital of Fudan University, Fudan University, Shanghai, 201102, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, 399 Wanyuan Road, Minhang, Shanghai, 201102, China
| | - F Bai
- National Children's Medical Center, Children's Hospital of Fudan University, Fudan University, Shanghai, 201102, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, 399 Wanyuan Road, Minhang, Shanghai, 201102, China
| | - Y Gui
- National Children's Medical Center, Children's Hospital of Fudan University, Fudan University, Shanghai, 201102, China.
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, 399 Wanyuan Road, Minhang, Shanghai, 201102, China.
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, 201102, China.
| |
Collapse
|
5
|
Shi DL. RNA-Binding Proteins as Critical Post-Transcriptional Regulators of Cardiac Regeneration. Int J Mol Sci 2023; 24:12004. [PMID: 37569379 PMCID: PMC10418649 DOI: 10.3390/ijms241512004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Myocardial injury causes death to cardiomyocytes and leads to heart failure. The adult mammalian heart has very limited regenerative capacity. However, the heart from early postnatal mammals and from adult lower vertebrates can fully regenerate after apical resection or myocardial infarction. Thus, it is of particular interest to decipher the mechanism underlying cardiac regeneration that preserves heart structure and function. RNA-binding proteins, as key regulators of post-transcriptional gene expression to coordinate cell differentiation and maintain tissue homeostasis, display dynamic expression in fetal and adult hearts. Accumulating evidence has demonstrated their importance for the survival and proliferation of cardiomyocytes following neonatal and postnatal cardiac injury. Functional studies suggest that RNA-binding proteins relay damage-stimulated cell extrinsic or intrinsic signals to regulate heart regenerative capacity by reprogramming multiple molecular and cellular processes, such as global protein synthesis, metabolic changes, hypertrophic growth, and cellular plasticity. Since manipulating the activity of RNA-binding proteins can improve the formation of new cardiomyocytes and extend the window of the cardiac regenerative capacity in mammals, they are potential targets of therapeutic interventions for cardiovascular disease. This review discusses our evolving understanding of RNA-binding proteins in regulating cardiac repair and regeneration, with the aim to identify important open questions that merit further investigations.
Collapse
Affiliation(s)
- De-Li Shi
- Department of Medical Research, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
- Laboratory of Developmental Biology (CNRS-UMR7622), Institute de Biologie Paris-Seine (IBPS), Sorbonne University, 75005 Paris, France
| |
Collapse
|
6
|
Lu J, Zhang C, Wang W, Xu W, Chen W, Tao L, Li Z, Zhang Y, Cheng J. Exposure to environmental concentrations of glyphosate induces cardiotoxicity through cellular senescence and reduced cell proliferation capacity. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 261:115112. [PMID: 37290295 DOI: 10.1016/j.ecoenv.2023.115112] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/01/2023] [Accepted: 06/04/2023] [Indexed: 06/10/2023]
Abstract
Glyphosate (GLY), the preeminent herbicide utilized globally, is known to be exposed to the environment and population on a chronic basis. Exposure to GLY and the consequent health risks are alarming public health problems that are attracting international attention. However, the cardiotoxicity of GLY has been a matter of dispute and uncertainty. Here, AC16 cardiomyocytes and zebrafish were exposed to GLY. This study found that low concentrations of GLY lead to morphological enlargement of AC16 human cardiomyocytes, indicating a senescent state. The increased expression of P16, P21, and P53 following exposure to GLY demonstrated that GLY causes senescence in AC16. Moreover, it was mechanistically confirmed that GLY-induced senescence in AC16 cardiomyocytes was produced by ROS-mediated DNA damage. In terms of in vivo cardiotoxicity, GLY decreased the proliferative capacity of cardiomyocytes in zebrafish through the notch signaling pathway, resulting in a reduction of cardiomyocytes. It was also found that GLY caused zebrafish cardiotoxicity associated with DNA damage and mitochondrial damage. KEGG analysis after RNA-seq shows a significant enrichment of protein processing pathways in the endoplasmic reticulum (ER) after GLY exposure. Importantly, GLY induced ER stress in AC16 cells and zebrafish by activating PERK-eIF2α-ATF4 pathway. Our study has thus provided the first novel insights into the mechanism underlying GLY-induced cardiotoxicity. Furthermore, our findings emphasize the need for increased attention to the potential cardiotoxic effects of GLY.
Collapse
Affiliation(s)
- Jian Lu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Cheng Zhang
- Department of Pathology,UT southwestern Medical Center, Dallas, TX 75390, United States
| | - Weiguo Wang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Wenping Xu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Weidong Chen
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Liming Tao
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Zhong Li
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yang Zhang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| | - Jiagao Cheng
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
7
|
Zhang C, Sun Y, Chen Z. An Efficient and Reproducible Method for the Isolation and Culture of Primary Cardiomyocytes from Adult Zebrafish. Zebrafish 2023. [PMID: 37262192 DOI: 10.1089/zeb.2023.0015] [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: 06/03/2023] Open
Abstract
Zebrafish is a popular animal model in regeneration studies due to their ability to regenerate the heart. Primary cardiomyocytes could be an alternative tool for studying the intrinsic mechanisms of cardiovascular disease in vitro. Thus, our objective is to develop an efficient protocol to isolate primary cardiomyocytes from zebrafish hearts. Low concentration of digestive enzyme (0.5 mg/mL collagenase type II) was utilized in our protocol to obtain single-cell suspension. The ventricles were fragmented, mechanically pipetted, and constantly shaken to ensure adequate contact between the tissues and the enzyme. Preplating the cell suspension onto culture plates for 2 h helped remove cardiac fibroblasts. The purity of isolated cells was validated by flow cytometry analysis of transgenic zebrafish with cardiomyocyte-specific expression of enhanced green fluorescent protein (EGFP) or endothelial cell-specific expression of mCherry. Quantitative real-time PCR analysis revealed a high level of the purity, with cardiac fibroblasts, endothelial cells, and epicardial cell markers scarcely detected in the purified cells. Altogether, this study established a reproducible protocol for isolating primary cardiomyocytes with high purity and activity from adult zebrafish hearts that can be cultured in vitro for up to 4 weeks. This protocol provides a valuable tool for studying the intrinsic mechanisms of cardiovascular disease in vitro using primary cardiomyocytes.
Collapse
Affiliation(s)
- Chunyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanyi Sun
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenyue Chen
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Mehdipour M, Park S, Huang GN. Unlocking cardiomyocyte renewal potential for myocardial regeneration therapy. J Mol Cell Cardiol 2023; 177:9-20. [PMID: 36801396 PMCID: PMC10699255 DOI: 10.1016/j.yjmcc.2023.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/28/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
Cardiovascular disease remains the leading cause of mortality worldwide. Cardiomyocytes are irreversibly lost due to cardiac ischemia secondary to disease. This leads to increased cardiac fibrosis, poor contractility, cardiac hypertrophy, and subsequent life-threatening heart failure. Adult mammalian hearts exhibit notoriously low regenerative potential, further compounding the calamities described above. Neonatal mammalian hearts, on the other hand, display robust regenerative capacities. Lower vertebrates such as zebrafish and salamanders retain the ability to replenish lost cardiomyocytes throughout life. It is critical to understand the varying mechanisms that are responsible for these differences in cardiac regeneration across phylogeny and ontogeny. Adult mammalian cardiomyocyte cell cycle arrest and polyploidization have been proposed as major barriers to heart regeneration. Here we review current models about why adult mammalian cardiac regenerative potential is lost including changes in environmental oxygen levels, acquisition of endothermy, complex immune system development, and possible cancer risk tradeoffs. We also discuss recent progress and highlight conflicting reports pertaining to extrinsic and intrinsic signaling pathways that control cardiomyocyte proliferation and polyploidization in growth and regeneration. Uncovering the physiological brakes of cardiac regeneration could illuminate novel molecular targets and offer promising therapeutic strategies to treat heart failure.
Collapse
Affiliation(s)
- Melod Mehdipour
- Cardiovascular Research Institute and 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; Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sangsoon Park
- Cardiovascular Research Institute and 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; Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Guo N Huang
- Cardiovascular Research Institute and 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; Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
9
|
Filosa A, Sawamiphak S. Heart development and regeneration-a multi-organ effort. FEBS J 2023; 290:913-930. [PMID: 34894086 DOI: 10.1111/febs.16319] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/22/2021] [Accepted: 12/10/2021] [Indexed: 12/15/2022]
Abstract
Development of the heart, from early morphogenesis to functional maturation, as well as maintenance of its homeostasis are tasks requiring collaborative efforts of cardiac tissue and different extra-cardiac organ systems. The brain, lymphoid organs, and gut are among the interaction partners that can communicate with the heart through a wide array of paracrine signals acting at local or systemic level. Disturbance of cardiac homeostasis following ischemic injury also needs immediate response from these distant organs. Our hearts replace dead muscles with non-contractile fibrotic scars. We have learned from animal models capable of scarless repair that regenerative capability of the heart does not depend only on competency of the myocardium and cardiac-intrinsic factors but also on long-range molecular signals originating in other parts of the body. Here, we provide an overview of inter-organ signals that take part in development and regeneration of the heart. We highlight recent findings and remaining questions. Finally, we discuss the potential of inter-organ modulatory approaches for possible therapeutic use.
Collapse
Affiliation(s)
- 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.,DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany
| |
Collapse
|
10
|
Future regenerative medicine developments and their therapeutic applications. Biomed Pharmacother 2023; 158:114131. [PMID: 36538861 DOI: 10.1016/j.biopha.2022.114131] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Although the currently available pharmacological assays can cure most pathological disorders, they have limited therapeutic value in relieving certain disorders like myocardial infarct, peripheral vascular disease, amputated limbs, or organ failure (e.g. renal failure). Pilot studies to overcome such problems using regenerative medicine (RM) delivered promising data. Comprehensive investigations of RM in zebrafish or reptilians are necessary for better understanding. However, the precise mechanisms remain poorly understood despite the tremendous amount of data obtained using the zebrafish model investigating the exact mechanisms behind their regenerative capability. Indeed, understanding such mechanisms and their application to humans can save millions of lives from dying due to potentially life-threatening events. Recent studies have launched a revolution in replacing damaged human organs via different approaches in the last few decades. The newly established branch of medicine (known as Regenerative Medicine aims to enhance natural repair mechanisms. This can be done through the application of several advanced broad-spectrum technologies such as organ transplantation, tissue engineering, and application of Scaffolds technology (support vascularization using an extracellular matrix), stem cell therapy, miRNA treatment, development of 3D mini-organs (organoids), and the construction of artificial tissues using nanomedicine and 3D bio-printers. Moreover, in the next few decades, revolutionary approaches in regenerative medicine will be applied based on artificial intelligence and wireless data exchange, soft intelligence biomaterials, nanorobotics, and even living robotics capable of self-repair. The present work presents a comprehensive overview that summarizes the new and future advances in the field of RM.
Collapse
|
11
|
Cardiac fibroblasts and mechanosensation in heart development, health and disease. Nat Rev Cardiol 2022; 20:309-324. [PMID: 36376437 DOI: 10.1038/s41569-022-00799-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/04/2022] [Indexed: 11/16/2022]
Abstract
The term 'mechanosensation' describes the capacity of cells to translate mechanical stimuli into the coordinated regulation of intracellular signals, cellular function, gene expression and epigenetic programming. This capacity is related not only to the sensitivity of the cells to tissue motion, but also to the decryption of tissue geometric arrangement and mechanical properties. The cardiac stroma, composed of fibroblasts, has been historically considered a mechanically passive component of the heart. However, the latest research suggests that the mechanical functions of these cells are an active and necessary component of the developmental biology programme of the heart that is involved in myocardial growth and homeostasis, and a crucial determinant of cardiac repair and disease. In this Review, we discuss the general concept of cell mechanosensation and force generation as potent regulators in heart development and pathology, and describe the integration of mechanical and biohumoral pathways predisposing the heart to fibrosis and failure. Next, we address the use of 3D culture systems to integrate tissue mechanics to mimic cardiac remodelling. Finally, we highlight the potential of mechanotherapeutic strategies, including pharmacological treatment and device-mediated left ventricular unloading, to reverse remodelling in the failing heart.
Collapse
|
12
|
de Sena-Tomás C, Aleman AG, Ford C, Varshney A, Yao D, Harrington JK, Saúde L, Ramialison M, Targoff KL. Activation of Nkx2.5 transcriptional program is required for adult myocardial repair. Nat Commun 2022; 13:2970. [PMID: 35624100 PMCID: PMC9142600 DOI: 10.1038/s41467-022-30468-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 05/03/2022] [Indexed: 11/09/2022] Open
Abstract
The cardiac developmental network has been associated with myocardial regenerative potential. However, the embryonic signals triggered following injury have yet to be fully elucidated. Nkx2.5 is a key causative transcription factor associated with human congenital heart disease and one of the earliest markers of cardiac progenitors, thus it serves as a promising candidate. Here, we show that cardiac-specific RNA-sequencing studies reveal a disrupted embryonic transcriptional profile in the adult Nkx2.5 loss-of-function myocardium. nkx2.5-/- fish exhibit an impaired ability to recover following ventricular apex amputation with diminished dedifferentiation and proliferation. Complex network analyses illuminate that Nkx2.5 is required to provoke proteolytic pathways necessary for sarcomere disassembly and to mount a proliferative response for cardiomyocyte renewal. Moreover, Nkx2.5 targets embedded in these distinct gene regulatory modules coordinate appropriate, multi-faceted injury responses. Altogether, our findings support a previously unrecognized, Nkx2.5-dependent regenerative circuit that invokes myocardial cell cycle re-entry, proteolysis, and mitochondrial metabolism to ensure effective regeneration in the teleost heart.
Collapse
Affiliation(s)
- Carmen de Sena-Tomás
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Angelika G Aleman
- Department of Physiology & Cellular Biophysics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Caitlin Ford
- Department of Genetics & Development, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Akriti Varshney
- Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Australian Regenerative Medicine Institute & Systems Biology Institute Australia, Monash University, Clayton, VIC, 3800, Australia
| | - Di Yao
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Jamie K Harrington
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Leonor Saúde
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Mirana Ramialison
- Australian Regenerative Medicine Institute & Systems Biology Institute Australia, Monash University, Clayton, VIC, 3800, Australia
- Murdoch Children's Research Institute & Department of Peadiatrics, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Kimara L Targoff
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA.
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
13
|
Grassinger JM, Henrich M, Echevarría AC, März I, Henrich E, Bartel A, Schneider M, Aupperle-Lellbach H. Correlation of Histopathological Changes in the Left Atrium and Left Atrial Appendage with the Degree of Dilation in Cats. J Comp Pathol 2021; 189:8-25. [PMID: 34886990 DOI: 10.1016/j.jcpa.2021.09.001] [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: 12/30/2020] [Revised: 04/12/2021] [Accepted: 09/16/2021] [Indexed: 10/20/2022]
Abstract
Although atrial dilation is a common finding in feline cardiac disease, detailed investigations of atrial pathology are rare in cats. The aim of the study was to investigate the correlation between pathohistological findings, morphometric data and the degree of dilation of the left atrial appendage (LAA) in 53 cats. Based on the LAA volume, the samples were grouped into normal/control (group 0, ≤1 ml [n = 9]), mildly dilated (group 1, >1 to ≤2 ml [n = 16]), moderately dilated (group 2, >2 to ≤3 ml [n = 14]) and markedly dilated (group 3, >3 ml [n = 14]) groups, independent of the underlying disease. Samples from the LAA and the left atrium (LA) were histologically evaluated using haematoxylin and eosin- and Picrosirius red-stained sections, and morphometrically analysed using an image analysis system. The degree of endo-, myo- and epicardial fibrosis was directly correlated with increased LAA dilation. Due to cardiomyocyte hyperplasia and hypertrophy, the mean thickness of the atrial wall was significantly greater in groups 1 and 2 than in group 0. Conversely, group 3 had a lower mean atrial wall thickness than groups 1 and 2, which was attributed to increased transmural fibrosis and cardiomyocyte atrophy. These findings reflect intensive dynamic remodelling processes during LA and LAA dilation, indicating that reversibility appears to be limited in cases of severe left atrial dilation.
Collapse
Affiliation(s)
- Julia M Grassinger
- LABOKLIN GmbH & Co. KG, Labor für Klinische Diagnostik, Bad Kissingen, Berlin, Germany.
| | - Manfred Henrich
- Institut für Veterinär-Pathologie, Veterinärmedizinische Fakultät der Justus-Liebig-Universität Gießen, Gießen, Berlin, Germany
| | - Argine C Echevarría
- LABOKLIN GmbH & Co. KG, Labor für Klinische Diagnostik, Bad Kissingen, Berlin, Germany
| | - Imke März
- Tierklinik Hofheim, Hofheim, Berlin, Germany
| | - Estelle Henrich
- Klinik für Kleintiere, Veterinärmedizinische Fakultät der Justus-Liebig-Universität Gießen, Gießen, Berlin, Germany
| | - Alexander Bartel
- Institut für Veterinär-Epidemiologie und Biometrie, Freie Universität Berlin, Berlin, Germany
| | - Matthias Schneider
- Klinik für Kleintiere, Veterinärmedizinische Fakultät der Justus-Liebig-Universität Gießen, Gießen, Berlin, Germany
| | | |
Collapse
|
14
|
Park H, Yun BH, Lim W, Song G. Dinitramine induces cardiotoxicity and morphological alterations on zebrafish embryo development. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 240:105982. [PMID: 34598048 DOI: 10.1016/j.aquatox.2021.105982] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/17/2021] [Accepted: 09/18/2021] [Indexed: 06/13/2023]
Abstract
Dinitramine (DN), an herbicide in the dinitroaniline family, is used in agricultural areas to prevent unwanted plant growth. Dinitroaniline herbicides inhibit cell division by preventing microtubulin synthesis. They are strongly absorbed by the soil and can contaminate groundwater; however, the mode of action of these herbicides in non-target organisms remains unclear. In this study, we examined the developmental toxicity of DN in zebrafish embryos exposed to 1.6, 3.2, and 6.4 mg/L DN, compared to embryos exposed to DMSO (control) for 96 h. Visual assessments using transgenic zebrafish (fli1:eGFP) indicated abnormal cardiac development with enlarged ventricles and atria, decreased heartbeats, and impaired cardiac function. Along with cardiac development, vessel formation and angiogenesis were suppressed through activation of the inflammatory response. In addition, exposure to 6.4 mg/L DN for 96 h induced cell death, with upregulation of genes related to apoptosis. Our results showed that DN induced morphological changes and triggered an inflammatory response and apoptotic cell death that can impair embryonic growth and survival, providing an important mechanism of DN in aquatic organisms.
Collapse
Affiliation(s)
- Hahyun Park
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Bo Hyun Yun
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Whasun Lim
- Department of Food and Nutrition, Kookmin University, Seoul, 02707, Republic of Korea.
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
15
|
Kirillova A, Han L, Liu H, Kühn B. Polyploid cardiomyocytes: implications for heart regeneration. Development 2021; 148:271050. [PMID: 34897388 DOI: 10.1242/dev.199401] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Terminally differentiated cells are generally thought to have arrived at their final form and function. Many terminally differentiated cell types are polyploid, i.e. they have multiple copies of the normally diploid genome. Mammalian heart muscle cells, termed cardiomyocytes, are one such example of polyploid cells. Terminally differentiated cardiomyocytes are bi- or multi-nucleated, or have polyploid nuclei. Recent mechanistic studies of polyploid cardiomyocytes indicate that they can limit cellular proliferation and, hence, heart regeneration. In this short Spotlight, we present the mechanisms generating bi- and multi-nucleated cardiomyocytes, and the mechanisms generating polyploid nuclei. Our aim is to develop hypotheses about how these mechanisms might relate to cardiomyocyte proliferation and cardiac regeneration. We also discuss how these new findings could be applied to advance cardiac regeneration research, and how they relate to studies of other polyploid cells, such as cancer cells.
Collapse
Affiliation(s)
- Anna Kirillova
- Medical Scientist Training Program, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, PA 15219, USA
| | - Lu Han
- Division of Cardiology, UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA.,Pediatric Institute for Heart Regeneration and Therapeutics (I-HRT), UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA
| | - Honghai Liu
- Division of Cardiology, UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA.,Pediatric Institute for Heart Regeneration and Therapeutics (I-HRT), UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA
| | - Bernhard Kühn
- Division of Cardiology, UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA.,Pediatric Institute for Heart Regeneration and Therapeutics (I-HRT), UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA.,McGowan Institute of Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| |
Collapse
|
16
|
Wang W, Hu YF, Pang M, Chang N, Yu C, Li Q, Xiong JW, Peng Y, Zhang R. BMP and Notch Signaling Pathways differentially regulate Cardiomyocyte Proliferation during Ventricle Regeneration. Int J Biol Sci 2021; 17:2157-2166. [PMID: 34239346 PMCID: PMC8241734 DOI: 10.7150/ijbs.59648] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/14/2021] [Indexed: 01/15/2023] Open
Abstract
Adult mammalian hearts show limited capacity to proliferate after injury, while zebrafish are capable to completely regenerate injured hearts through the proliferation of spared cardiomyocytes. BMP and Notch signaling pathways have been implicated in cardiomyocyte proliferation during zebrafish heart regeneration. However, the molecular mechanism underneath this process as well as the interaction between these two pathways remains to be further explored. In this study we showed BMP signaling was activated after ventricle ablation and acted epistatic downstream of Notch signaling. Inhibition of both signaling pathways differentially influenced ventricle regeneration and cardiomyocyte proliferation, as revealed by time-lapse analysis using a cardiomyocyte-specific FUCCI (fluorescent ubiquitylation-based cell cycle indicator) system. Further experiments revealed that inhibition of BMP and Notch signaling led to cell-cycle arrest at different phases. Overall, our results shed light on the interaction between BMP and Notch signaling pathways and their functions in cardiomyocyte proliferation during cardiac regeneration.
Collapse
Affiliation(s)
- Wenyuan Wang
- School of Life Sciences, Fudan University, Shanghai, China
| | - Ye-Fan Hu
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Meijun Pang
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Nannan Chang
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Chunxiao Yu
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Qi Li
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jing-Wei Xiong
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Yuanyuan Peng
- School of Life Sciences, Fudan University, Shanghai, China
| | - Ruilin Zhang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
17
|
De Novo Transcriptome Profiling of Brain Tissue from the Annual Killifish Nothobranchius guentheri. Life (Basel) 2021; 11:life11020137. [PMID: 33670176 PMCID: PMC7916979 DOI: 10.3390/life11020137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/13/2022] Open
Abstract
Nothobranchius is a genus of small annual killifish found in Africa. Due to the relatively short lifespan, as well as easy breeding and care, Nothobranchius fish are becoming widely used as a vertebrate model system. Studying the genome and transcriptome of these fish is essential for advancing the field. In this study, we performed de novo transcriptome assembly of brain tissues from Nothobranchius guentheri using Trinity. Annotation of 104,271 potential genes (with transcripts longer than 500 bp) was carried out; for 24,967 genes (53,654 transcripts), in which at least one GO annotation was derived. We also analyzed the effect of a long-term food supplement with Torin 2, second-generation ATP-competitive inhibitor of mTOR, on the gene expression changes in brain tissue of adult N. guentheri. Overall, 1491 genes in females and 249 genes in males were differently expressed under Torin 2-supplemented diet. According to the Gene Set Enrichment Analysis (GSEA), the majority of identified genes were predominantly involved in the regulation of metabolic process, dendritic spine maintenance, circadian rhythms, retrotransposition, and immune response. Thus, we have provided the first transcriptome assembly and assessed the differential gene expression in response to exposure to Torin 2, which allow a better understanding of molecular changes in the brain tissues of adult fish in the mTOR pathway inhibition.
Collapse
|
18
|
Helston O, Amaya E. Reactive oxygen species during heart regeneration in zebrafish: Lessons for future clinical therapies. Wound Repair Regen 2021; 29:211-224. [PMID: 33471940 PMCID: PMC8611801 DOI: 10.1111/wrr.12892] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/30/2020] [Accepted: 12/22/2020] [Indexed: 11/30/2022]
Abstract
In humans, myocardial infarction (MI) is associated with irreversible damage to heart tissue, resulting in increased morbidity and mortality in patients. By comparison, the zebrafish (Danio rerio) is capable of repairing damaged and injured hearts by activating a full regenerative response. By studying model organisms that can regenerate loss heart tissue following injury, such as the zebrafish, a greater insight will be gained into the molecular pathways that can induce and sustain a regenerative response following injury. There is hope that such information may lead to new treatments or therapies aimed at stimulating a better regenerative response in humans that have suffered heart attacks. Recent findings in zebrafish have highlighted an important role for sustained elevated levels of Reactive Oxygen Species (ROS), including hydrogen peroxide (H2O2) in the promotion of a regenerative response. Given that elevated levels of H2O2 can be harmful, simply elevating ROS levels directly may not be easy or practical to translate clinically. An alternative approach would be to identify the critical downstream targets of ROS in the promotion of heart regeneration, and then target these clinically using drugs. One such family of potential downstream targets of ROS during heart regeneration are the family of protein tyrosine phosphatases (PTPs), which are known to be exquisitely sensitive to redox regulation and whose inhibition have been linked to the promotion of heart regeneration in zebrafish. In this review, we present an overview of the zebrafish as a model organism for studying cardiac regeneration, including the molecular mechanisms by which cardiac regeneration occurs in response to injury. We then present recent findings linking elevated ROS levels to heart regeneration and their potential downstream targets, the PTPs, including protein tyrosine phosphatase 1B (PTP1B) and the dual specificity phosphatase 6 (DUSP6) in the promotion of heart regeneration.
Collapse
Affiliation(s)
- Olivia Helston
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Enrique Amaya
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
19
|
Chen J. NF-Y is critical for the proper growth of zebrafish embryonic heart and its cardiomyocyte proliferation. Genesis 2021; 59:e23408. [PMID: 33417743 DOI: 10.1002/dvg.23408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 12/27/2020] [Accepted: 12/29/2020] [Indexed: 11/06/2022]
Abstract
The ubiquitous NF-Y gene regulates the expression of different genes in various signaling pathways. However, the function of NF-Y in zebrafish heart development is largely unknown. Previously we identified a same group of cell cycle related gene cluster (CCRG) was downregulated in the embryonic hearts with impeded growth due to various stresses. The promoter regions of these CCRG genes shared a most common motif for NF-Y. Chromatin immunoprecipitation experiment demonstrated that the binding of NF-Y to its motif was real on the CCRG candidate gene promoters. Knockdown of embryonic NF-Y by morpholinos led to a small heart, mimicking the abnormal heart phenotype caused by other stresses. In parallel the expression of certain CCRG candidate genes was reduced in the NF-Y A morphant hearts exposed to malignant environments. Absence of NF-Y A also led to undermine cardiomyocyte proliferation and hence less total number of caridomyocytes per heart. Trans-AM Elisa experiment also found that in the presence of the stresses such as TCDD and TNNT2 MO, the binding capacity of NF-Y A subunit to its core motif was reduced. We conclude that NF-Y sustains proper cardiomyocyte proliferation in the heart, thus it plays a positive role in promoting early zebrafish heart growth.
Collapse
Affiliation(s)
- Jing Chen
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, College of Life Sciences, Fujian Normal University, Fuzhou, China
| |
Collapse
|
20
|
Mousavi A, Vahdat S, Baheiraei N, Razavi M, Norahan MH, Baharvand H. Multifunctional Conductive Biomaterials as Promising Platforms for Cardiac Tissue Engineering. ACS Biomater Sci Eng 2020; 7:55-82. [PMID: 33320525 DOI: 10.1021/acsbiomaterials.0c01422] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Adult cardiomyocytes are terminally differentiated cells that result in minimal intrinsic potential for the heart to self-regenerate. The introduction of novel approaches in cardiac tissue engineering aims to repair damages from cardiovascular diseases. Recently, conductive biomaterials such as carbon- and gold-based nanomaterials, conductive polymers, and ceramics that have outstanding electrical conductivity, acceptable mechanical properties, and promoted cell-cell signaling transduction have attracted attention for use in cardiac tissue engineering. Nevertheless, comprehensive classification of conductive biomaterials from the perspective of cardiac cell function is a subject for discussion. In the present review, we classify and summarize the unique properties of conductive biomaterials considered beneficial for cardiac tissue engineering. We attempt to cover recent advances in conductive biomaterials with a particular focus on their effects on cardiac cell functions and proposed mechanisms of action. Finally, current problems, limitations, challenges, and suggested solutions for applications of these biomaterials are presented.
Collapse
Affiliation(s)
- Ali Mousavi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Sadaf Vahdat
- Tissue Engineering and Applied Cell Sciences Division, Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, 14117-13116 Tehran, Iran.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, 1665659911 Tehran, Iran
| | - Nafiseh Baheiraei
- Tissue Engineering and Applied Cell Sciences Division, Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, 14117-13116 Tehran, Iran
| | - Mehdi Razavi
- Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, Florida 32816, United States
| | - Mohammad Hadi Norahan
- Centro de Biotecnología-FEMSA, Department of Sciences, Tecnologico de Monterrey, Monterrey 64849, NL, México
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, 1665659911 Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
21
|
Ng DCH, Richards DK, Mills RJ, Ho UY, Perks HL, Tucker CR, Voges HK, Pagan JK, Hudson JE. Centrosome Reduction Promotes Terminal Differentiation of Human Cardiomyocytes. Stem Cell Reports 2020; 15:817-826. [PMID: 32946803 PMCID: PMC7561510 DOI: 10.1016/j.stemcr.2020.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 08/18/2020] [Accepted: 08/18/2020] [Indexed: 01/10/2023] Open
Abstract
Centrosome reduction and redistribution of pericentriolar material (PCM) coincides with cardiomyocyte transitions to a post-mitotic and matured state. However, it is unclear whether centrosome changes are a cause or consequence of terminal differentiation. We validated that centrosomes were intact and functional in proliferative human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs), consistent with their immature phenotype. We generated acentrosomal hPSC-CMs, through pharmacological inhibition of centriole duplication, and showed that centrosome loss was sufficient to promote post-mitotic transitions and aspects of cardiomyocyte maturation. As Hippo kinases are activated during post-natal cardiac maturation, we pharmacologically activated the Hippo pathway using C19, which was sufficient to trigger centrosome disassembly and relocalization of PCM components to perinuclear membranes. This was due to specific activation of Hippo kinases, as direct inhibition of YAP-TEAD interactions with verteporfin had no effect on centrosome organization. This suggests that Hippo kinase-centrosome remodeling may play a direct role in cardiac maturation. Centrosomes are intact and functional in immature human cardiomyocytes Centrosome loss promotes maturation of human cardiomyocytes Centrosomes are returned with cell cycle reinitiation in post-natal cardiomyocytes Hippo kinases promote disassembly and redistribution of centrosomal proteins
Collapse
Affiliation(s)
- Dominic C H Ng
- School of Biomedical Sciences, University of Queensland, Saint Lucia, QLD, Australia.
| | - Dominic K Richards
- School of Biomedical Sciences, University of Queensland, Saint Lucia, QLD, Australia
| | - Richard J Mills
- Cardiac Bioengineering Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Uda Y Ho
- School of Biomedical Sciences, University of Queensland, Saint Lucia, QLD, Australia
| | - Hannah L Perks
- School of Biomedical Sciences, University of Queensland, Saint Lucia, QLD, Australia
| | - Callum R Tucker
- School of Biomedical Sciences, University of Queensland, Saint Lucia, QLD, Australia
| | - Holly K Voges
- Cardiac Bioengineering Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Julia K Pagan
- School of Biomedical Sciences, University of Queensland, Saint Lucia, QLD, Australia
| | - James E Hudson
- Cardiac Bioengineering Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
22
|
Zhu F, Meng Q, Yu Y, Shao L, Shen Z. Adult Cardiomyocyte Proliferation: a New Insight for Myocardial Infarction Therapy. J Cardiovasc Transl Res 2020; 14:457-466. [PMID: 32820393 DOI: 10.1007/s12265-020-10067-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/10/2020] [Indexed: 12/20/2022]
Abstract
Myocardial infarction leads to cardiomyocyte loss, ensuing ventricular pathological remodeling, dramatic impairment of cardiac function, and ultimately heart failure. Unfortunately, the existing therapeutical treatments cannot directly replenish the lost myocytes in the injured myocardium and the long-term prognosis of heart failure after myocardial infarction remains poor. Growing investigations have demonstrated that the adult mammalian cardiomyocytes possess very limited proliferation capacity, and that was not enough to restore the injured heart. Recently, many studies were targeting to promote cardiomyocyte proliferation via inducing cardiomyocyte cell cycle re-entry for cardiac repair after myocardial infarction. Indeed, these results showed it is a feasible way to stimulate terminally differentiated cardiomyocyte proliferation. Here, we reviewed the major mechanisms and the potential targets for stimulating mammalian adult cardiomyocyte proliferation specifically. This will provide a new therapeutic strategy for the clinical treatment of myocardial infarction by activating the endogenous regeneration. Graphical abstract.
Collapse
Affiliation(s)
- Feng Zhu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Qingyou Meng
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
| | - You Yu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Lianbo Shao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China.
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
23
|
Velayutham N, Alfieri CM, Agnew EJ, Riggs KW, Baker RS, Ponny SR, Zafar F, Yutzey KE. Cardiomyocyte cell cycling, maturation, and growth by multinucleation in postnatal swine. J Mol Cell Cardiol 2020; 146:95-108. [PMID: 32710980 DOI: 10.1016/j.yjmcc.2020.07.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/19/2020] [Accepted: 07/07/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Rodent cardiomyocytes (CM) undergo mitotic arrest and decline of mononucleated-diploid population post-birth, which are implicated in neonatal loss of heart regenerative potential. However, the dynamics of postnatal CM maturation are largely unknown in swine, despite a similar neonatal cardiac regenerative capacity as rodents. Here, we provide a comprehensive analysis of postnatal cardiac maturation in swine, including CM cell cycling, multinucleation and hypertrophic growth, as well as non-CM cardiac factors such as extracellular matrix (ECM), immune cells, capillaries, and neurons. Our study reveals discordance in postnatal pig heart maturational events compared to rodents. METHODS AND RESULTS Left-ventricular myocardium from White Yorkshire-Landrace pigs at postnatal day (P)0 to 6 months (6mo) was analyzed. Mature cardiac sarcomeric characteristics, such as fetal TNNI1 repression and Cx43 co-localization to cell junctions, were not evident until P30 in pigs. In CMs, appreciable binucleation is observed by P7, with extensive multinucleation (4-16 nuclei per CM) beyond P15. Individual CM nuclei remain predominantly diploid at all ages. CM mononucleation at ~50% incidence is observed at P7-P15, and CM mitotic activity is measurable up to 2mo. CM cross-sectional area does not increase until 2mo-6mo in pigs, though longitudinal CM growth proportional to multinucleation occurs after P15. RNAseq analysis of neonatal pig left ventricles showed increased expression of ECM maturation, immune signaling, neuronal remodeling, and reactive oxygen species response genes, highlighting significance of the non-CM milieu in postnatal mammalian heart maturation. CONCLUSIONS CM maturational events such as decline of mononucleation and cell cycle arrest occur over a 2-month postnatal period in pigs, despite reported loss of heart regenerative potential by P3. Moreover, CMs grow primarily by multinucleation and longitudinal hypertrophy in older pig CMs, distinct from mice and humans. These differences are important to consider for preclinical testing of cardiovascular therapies using swine, and may offer opportunities to study aspects of heart regeneration unavailable in other models.
Collapse
Affiliation(s)
- Nivedhitha Velayutham
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Molecular and Developmental Biology Graduate Program, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Christina M Alfieri
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Emma J Agnew
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kyle W Riggs
- Division of Pediatric Cardiothoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - R Scott Baker
- Division of Pediatric Cardiothoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sithara Raju Ponny
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Farhan Zafar
- Division of Pediatric Cardiothoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Katherine E Yutzey
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Molecular and Developmental Biology Graduate Program, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
24
|
Budine TE, de Sena-Tomás C, Williams MLK, Sepich DS, Targoff KL, Solnica-Krezel L. Gon4l/Udu regulates cardiomyocyte proliferation and maintenance of ventricular chamber identity during zebrafish development. Dev Biol 2020; 462:223-234. [PMID: 32272116 PMCID: PMC10318589 DOI: 10.1016/j.ydbio.2020.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 01/26/2020] [Accepted: 03/02/2020] [Indexed: 01/03/2023]
Abstract
Vertebrate heart development requires spatiotemporal regulation of gene expression to specify cardiomyocytes, increase the cardiomyocyte population through proliferation, and to establish and maintain atrial and ventricular cardiac chamber identities. The evolutionarily conserved chromatin factor Gon4-like (Gon4l), encoded by the zebrafish ugly duckling (udu) locus, has previously been implicated in cell proliferation, cell survival, and specification of mesoderm-derived tissues including blood and somites, but its role in heart formation has not been studied. Here we report two distinct roles of Gon4l/Udu in heart development: regulation of cell proliferation and maintenance of ventricular identity. We show that zygotic loss of udu expression causes a significant reduction in cardiomyocyte number at one day post fertilization that becomes exacerbated during later development. We present evidence that the cardiomyocyte deficiency in udu mutants results from reduced cell proliferation, unlike hematopoietic deficiencies attributed to TP53-dependent apoptosis. We also demonstrate that expression of the G1/S-phase cell cycle regulator, cyclin E2 (ccne2), is reduced in udu mutant hearts, and that the Gon4l protein associates with regulatory regions of the ccne2 gene during early embryogenesis. Furthermore, udu mutant hearts exhibit a decrease in the proportion of ventricular cardiomyocytes compared to atrial cardiomyocytes, concomitant with progressive reduction of nkx2.5 expression. We further demonstrate that udu and nkx2.5 interact to maintain the proportion of ventricular cardiomyocytes during development. However, we find that ectopic expression of nkx2.5 is not sufficient to restore ventricular chamber identity suggesting that Gon4l regulates cardiac chamber patterning via multiple pathways. Together, our findings define a novel role for zygotically-expressed Gon4l in coordinating cardiomyocyte proliferation and chamber identity maintenance during cardiac development.
Collapse
Affiliation(s)
- Terin E Budine
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carmen de Sena-Tomás
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Margot L K Williams
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Diane S Sepich
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kimara L Targoff
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Lila Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
25
|
Wasserman AH, Venkatesan M, Aguirre A. Bioactive Lipid Signaling in Cardiovascular Disease, Development, and Regeneration. Cells 2020; 9:E1391. [PMID: 32503253 PMCID: PMC7349721 DOI: 10.3390/cells9061391] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/23/2020] [Accepted: 06/01/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD) remains a leading cause of death globally. Understanding and characterizing the biochemical context of the cardiovascular system in health and disease is a necessary preliminary step for developing novel therapeutic strategies aimed at restoring cardiovascular function. Bioactive lipids are a class of dietary-dependent, chemically heterogeneous lipids with potent biological signaling functions. They have been intensively studied for their roles in immunity, inflammation, and reproduction, among others. Recent advances in liquid chromatography-mass spectrometry techniques have revealed a staggering number of novel bioactive lipids, most of them unknown or very poorly characterized in a biological context. Some of these new bioactive lipids play important roles in cardiovascular biology, including development, inflammation, regeneration, stem cell differentiation, and regulation of cell proliferation. Identifying the lipid signaling pathways underlying these effects and uncovering their novel biological functions could pave the way for new therapeutic strategies aimed at CVD and cardiovascular regeneration.
Collapse
Affiliation(s)
- Aaron H. Wasserman
- Regenerative Biology and Cell Reprogramming Laboratory, Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA; (A.H.W.); (M.V.)
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Manigandan Venkatesan
- Regenerative Biology and Cell Reprogramming Laboratory, Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA; (A.H.W.); (M.V.)
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Aitor Aguirre
- Regenerative Biology and Cell Reprogramming Laboratory, Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI 48824, USA; (A.H.W.); (M.V.)
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
26
|
He JQ, Barron C. Signaling pathways in modulation of tissue and organ regeneration in vertebrates. Semin Cell Dev Biol 2020; 100:1-2. [PMID: 31831358 PMCID: PMC9793442 DOI: 10.1016/j.semcdb.2019.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
27
|
Abstract
Heart failure is a major cause of death worldwide owing to the inability of the adult human heart to regenerate after a heart attack. However, many vertebrate species are capable of complete cardiac regeneration following injury. In this Review, we discuss the various model organisms of cardiac regeneration, and outline what they have taught us thus far about the cellular and molecular responses essential for optimal cardiac repair. We compare across different species, highlighting evolutionarily conserved mechanisms of regeneration and demonstrating the importance of developmental gene expression programmes, plasticity of the heart and the pathophysiological environment for the regenerative response. Additionally, we discuss how the findings from these studies have led to improvements in cardiac repair in preclinical models such as adult mice and pigs, and discuss the potential to translate these findings into therapeutic approaches for human patients following myocardial infarction.
Collapse
Affiliation(s)
- Eleanor L Price
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Joaquim M Vieira
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Paul R Riley
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| |
Collapse
|
28
|
Beffagna G. Zebrafish as a Smart Model to Understand Regeneration After Heart Injury: How Fish Could Help Humans. Front Cardiovasc Med 2019; 6:107. [PMID: 31448289 PMCID: PMC6691037 DOI: 10.3389/fcvm.2019.00107] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/19/2019] [Indexed: 12/26/2022] Open
Abstract
Myocardial infarction (MI) in humans is a common cause of cardiac injury and results in irreversible loss of myocardial cells and formation of fibrotic scar tissue. This fibrotic tissue preserves the integrity of the ventricular wall but undermines pump function, leading to congestive heart failure. Unfortunately, the mammalian heart is unable to replace cardiomyocytes, so the life expectancy for patients after an episode of MI is lower than for most common types of cancers. Whereas, humans cannot efficiently regenerate their heart after injury, the teleost zebrafish have the capability to repair a “broken” heart. The zebrafish is probably one of the most important models for developmental and regenerative biology of the heart. In the last decades, the zebrafish has become increasingly important for scientific research: it has many characteristics that make it a smart model for studying human disease. Moreover, adult zebrafish efficiently regenerate their hearts following different forms of injury. Due to these characteristics, and to the availability of genetic approaches, and biosensor zebrafish lines, it has been established useful for studying molecular mechanisms of heart regeneration. Regeneration of cardiomyocytes in zebrafish is not based on stem cells or transdifferentiation of other cells but on the proliferation of preexisting cardiomyocytes. For this reason, future studies into the zebrafish cardiac regenerative mechanisms could identify specific molecules able to regulate the proliferation of preexisting cardiomyocytes; these factors may be studied in order to understand regulation of myocardial plasticity in cardiac repair processes after injury and, in particular, after MI in humans.
Collapse
Affiliation(s)
- Giorgia Beffagna
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Padua, Italy
| |
Collapse
|
29
|
Landim-Vieira M, Schipper JM, Pinto JR, Chase PB. Cardiomyocyte nuclearity and ploidy: when is double trouble? J Muscle Res Cell Motil 2019; 41:329-340. [PMID: 31317457 DOI: 10.1007/s10974-019-09545-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/12/2019] [Indexed: 01/23/2023]
Abstract
Considerable effort has gone into investigating mechanisms that underlie the developmental transition in which mammalian cardiomyocytes (CMs) switch from being able to proliferate during development, to essentially having lost that ability at maturity. This problem is interesting not only for scientific curiosity, but also for its clinical relevance because controlling the ability of mature CMs to replicate would provide a much-needed approach for restoring cardiac function in damaged hearts. In this review, we focus on the propensity of mature mammalian CMs to be multinucleated and polyploid, and the extent to which this may be necessary for normal physiology yet possibly disadvantageous in some circumstances. In this context, we explore whether the concept of the myonuclear domain (MND) in multinucleated skeletal muscle fibers might apply to cardiomyocytes, and whether cardio-MND size might be related to the transition of CMs to become multinuclear. Nuclei in CMs are almost certainly integrators of not only biochemical, but also-because of their central location within the myofibrils-mechanical information, and this multimodal, integrative function in adult CMs-involving molecules that have been extensively studied along with newly identified possibilities-could influence both gene expression as well as replication of the genome and the nuclei themselves.
Collapse
Affiliation(s)
- Maicon Landim-Vieira
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Joslyn M Schipper
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA.,Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - J Renato Pinto
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - P Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL, USA. .,Department of Biological Science, Florida State University, Biology Unit One Room 206, 81 Chieftain Way, Tallahassee, FL, 32306-4370, USA.
| |
Collapse
|
30
|
Vilches-Moure JG. Embryonic Chicken ( Gallus gallus domesticus) as a Model of Cardiac Biology and Development. Comp Med 2019; 69:184-203. [PMID: 31182184 PMCID: PMC6591676 DOI: 10.30802/aalas-cm-18-000061] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/06/2018] [Accepted: 11/29/2018] [Indexed: 12/13/2022]
Abstract
Cardiovascular disease remains one of the top contributors to morbidity and mortality in the United States. Increasing evidence suggests that many processes, pathways, and programs observed during development and organogenesis are recapitulated in adults in the face of disease. Therefore, a heightened understanding of cardiac development and organogenesis will help increase our understanding of developmental defects and cardiovascular diseases in adults. Chicks have long served as a model system in which to study developmental problems. Detailed descriptions of morphogenesis, low cost, accessibility, ease of manipulation, and the optimization of genetic engineering techniques have made chicks a robust model for studying development and make it a powerful platform for cardiovascular research. This review summarizes the cardiac developmental milestones of embryonic chickens, practical considerations when working with chicken embryos, and techniques available for use in chicks (including tissue chimeras, genetic manipulations, and live imaging). In addition, this article highlights examples that accentuate the utility of the embryonic chicken as model system in which to study cardiac development, particularly epicardial development, and that underscore the importance of how studying development informs our understanding of disease.
Collapse
Affiliation(s)
- José G Vilches-Moure
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, California,
| |
Collapse
|
31
|
Hirose K, Payumo AY, Cutie S, Hoang A, Zhang H, Guyot R, Lunn D, Bigley RB, Yu H, Wang J, Smith M, Gillett E, Muroy SE, Schmid T, Wilson E, Field KA, Reeder DM, Maden M, Yartsev MM, Wolfgang MJ, Grützner F, Scanlan TS, Szweda LI, Buffenstein R, Hu G, Flamant F, Olgin JE, Huang GN. Evidence for hormonal control of heart regenerative capacity during endothermy acquisition. Science 2019; 364:184-188. [PMID: 30846611 DOI: 10.1126/science.aar2038] [Citation(s) in RCA: 246] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/15/2018] [Accepted: 02/21/2019] [Indexed: 12/11/2022]
Abstract
Tissue regenerative potential displays striking divergence across phylogeny and ontogeny, but the underlying mechanisms remain enigmatic. Loss of mammalian cardiac regenerative potential correlates with cardiomyocyte cell-cycle arrest and polyploidization as well as the development of postnatal endothermy. We reveal that diploid cardiomyocyte abundance across 41 species conforms to Kleiber's law-the ¾-power law scaling of metabolism with bodyweight-and inversely correlates with standard metabolic rate, body temperature, and serum thyroxine level. Inactivation of thyroid hormone signaling reduces mouse cardiomyocyte polyploidization, delays cell-cycle exit, and retains cardiac regenerative potential in adults. Conversely, exogenous thyroid hormones inhibit zebrafish heart regeneration. Thus, our findings suggest that loss of heart regenerative capacity in adult mammals is triggered by increasing thyroid hormones and may be a trade-off for the acquisition of endothermy.
Collapse
Affiliation(s)
- Kentaro Hirose
- Cardiovascular Research Institute and 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
- Cardiovascular Research Institute and 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
| | - Stephen Cutie
- Cardiovascular Research Institute and 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
| | - Alison Hoang
- Cardiovascular Research Institute and 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
| | - Hao Zhang
- Department of Medicine, Division of Cardiology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Romain Guyot
- Department of Internal Medicine, Institut de Génomique Fonctionnelle de Lyon, Institut National de la Recherche Agronomique, Université Lyon 1, CNRS, École Normale Superieure de Lyon, 69 007 France
| | - Dominic Lunn
- Cardiovascular Research Institute and 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
| | - Rachel B Bigley
- Cardiovascular Research Institute and 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
| | - Hongyao Yu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Jiajia Wang
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Megan Smith
- Calico Life Sciences, 1170 Veterans Boulevard, South San Francisco, CA 94080, USA
| | - Ellen Gillett
- School of Biological Sciences, University of Adelaide, South Australia, Adelaide 5005, Australia
| | - Sandra E Muroy
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94708, USA
| | - Tobias Schmid
- Helen Wills Neuroscience Institute and Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94708, USA
| | - Emily Wilson
- Department of Medicine, Division of Cardiology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Kenneth A Field
- Department of Biology, Bucknell University, Lewisburg, PA 17837, USA
| | - DeeAnn M Reeder
- Department of Biology, Bucknell University, Lewisburg, PA 17837, USA
| | - Malcom Maden
- Department of Biology and UF Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| | - Michael M Yartsev
- Helen Wills Neuroscience Institute and Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94708, USA
| | - Michael J Wolfgang
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Frank Grützner
- School of Biological Sciences, University of Adelaide, South Australia, Adelaide 5005, Australia
| | - Thomas S Scanlan
- Department of Physiology and Pharmacology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Luke I Szweda
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Rochelle Buffenstein
- Calico Life Sciences, 1170 Veterans Boulevard, South San Francisco, CA 94080, USA
| | - Guang Hu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Frederic Flamant
- Department of Internal Medicine, Institut de Génomique Fonctionnelle de Lyon, Institut National de la Recherche Agronomique, Université Lyon 1, CNRS, École Normale Superieure de Lyon, 69 007 France
| | - Jeffrey E Olgin
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA.,Department of Medicine, Division of Cardiology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Guo N Huang
- Cardiovascular Research Institute and 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
| |
Collapse
|
32
|
Rastgar S, Alijani Ardeshir R, Movahedinia A, Zabihi E, Salati AP, Salamat N. Spontaneously contracting cell aggregates derived from grass carp heart as a promising tool in in vitro heart research. Cytotechnology 2019; 71:261-266. [PMID: 30600462 DOI: 10.1007/s10616-018-0281-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 11/09/2018] [Indexed: 12/15/2022] Open
Abstract
Regarding challenges in isolation and maintenance of cultured heart cells, introduction of new in vitro heart model that is stable and exhibits long-term spontaneously contracting cell aggregates (SCCs), whose electrophysiological properties are comparable to the human heart, is required. This research aimed to establish cardiac primary cells and to evaluate the effects of culture conditions. Also the effect of fish age on beating SCC and cardiac cell morphology were studied. Twelve healthy grass carps (Ctenopharyngodon idella) were divided into four groups based on their age. Non-enzymatic explant culture was used and heart explants were incubated at 21-31 °C for 60 days. After proliferation of the cardiac primary cells, changes in their morphology and their beatings were recorded. The results showed that the explants derived from different age of grass carp fish are fully viable and proliferative with formation of SCC for a long period of time. By increasing the number of adhered cells, the cardiac primary cells became more flat and elongated. Increasing the medium temperature and fetal bovine serum concentration in culture medium led to decline and enhancement in beat frequencies of heart cell aggregates, respectively. Also, grass carp heart explant had high potential in regeneration (especially in young fish) and thus high feasibility to generate stable long-term cultures. In general, it seems that explant culture of heart from grass carp can be considered as a promising tool in heart research area.
Collapse
Affiliation(s)
- Sara Rastgar
- Department of Marine Biology, Faculty of Marine Sciences, Khorramshahr University of Marine Science and Technology, P.O. Box 669, Khorramshahr, Iran
| | - Rashid Alijani Ardeshir
- Department of Marine Biology, Faculty of Marine Sciences, Khorramshahr University of Marine Science and Technology, P.O. Box 669, Khorramshahr, Iran
| | - Abdolali Movahedinia
- Department of Marine Biology, Faculty of Marine Sciences, Khorramshahr University of Marine Science and Technology, P.O. Box 669, Khorramshahr, Iran.,Department of Marine Biology, Faculty of Marine Sciences, University of Mazandaran, Babolsar, Iran
| | - Ebrahim Zabihi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| | - Amir Parviz Salati
- Department of Fisheries, Faculty of Marine Natural Resources, Khorramshahr University of Marine Science and Technology, P.O. Box 669, Khorramshahr, Iran
| | - Negin Salamat
- Department of Marine Biology, Faculty of Marine Sciences, Khorramshahr University of Marine Science and Technology, P.O. Box 669, Khorramshahr, Iran
| |
Collapse
|
33
|
Rosa M, Alfonsina G, Sandra I, Loubna B, Serena L, Yamine MB, Mariacristina F, Carmine R, Tommaso A, Youssef A, Carmela CM. Selenoprotein T as a new positive inotrope in the goldfish Carassius auratus. J Exp Biol 2019; 222:jeb.201202. [DOI: 10.1242/jeb.201202] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/08/2019] [Indexed: 12/14/2022]
Abstract
Selenoprotein T (SELENOT) is a thioredoxin-like protein, which mediates oxidoreductase functions via its redox active motif Cys-X-X-Sec. In mammals, SELENOT is expressed during ontogenesis and progressively decreases in adult tissues. In the heart, it is re-expressed after ischemia and induces cardioprotection against ischemia/reperfusion (I/R) injury. SELENOT is present in teleost fish, including the goldfish Carassius auratus. This study aimed to evaluate the cardiac expression of SELENOT, and the effects of exogenous PSELT (a 43-52 SELENOT derived-peptide) on the heart function of C. auratus, a hypoxia tolerance fish model. We found that SELENOT was expressed in cardiac extracts of juvenile and adult fish, located in the sarcoplasmic reticulum (SR) together with calsequestrin-2. Expression increased under acute hypoxia. On ex vivo isolated and perfused goldfish heart preparations, under normoxia, PSELT dose-dependently increased Stroke Volume (SV), Cardiac Output (Q̇), and Stroke Work (SW), by involving cAMP, PKA, L-type calcium channels, SERCA2a pumps, and pAkt. Under hypoxia, PSELT did not affect myocardial contractility. Only at higher concentrations (10−8 -10−7 M) an increase of SV and Q̇ was observed. It also reduced the cardiac expression of 3-NT, a tissue marker of nitrosative stress which increases under low oxygen availability. These data are the first to propose SELENOT 43-52, PSELT, as a cardiac modulator in fish, with a potential protective role under hypoxia.
Collapse
Affiliation(s)
- Mazza Rosa
- Laboratory of Organ and System Physiology, Dept. of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Gattuso Alfonsina
- Laboratory of Organ and System Physiology, Dept. of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Imbrogno Sandra
- Laboratory of Organ and System Physiology, Dept. of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Boukhzar Loubna
- Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie and Centre Universitaire de Recherche et D'Innovation en Biologie, Normandie University, UNIROUEN, INSERM, Rouen, France
| | - Leo Serena
- Laboratory of Organ and System Physiology, Dept. of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Mallouki Ben Yamine
- Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie and Centre Universitaire de Recherche et D'Innovation en Biologie, Normandie University, UNIROUEN, INSERM, Rouen, France
| | - Filice Mariacristina
- Laboratory of Organ and System Physiology, Dept. of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Rocca Carmine
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Dept. of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Angelone Tommaso
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Dept. of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Anouar Youssef
- Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine, Institut de Recherche et d'Innovation Biomédicale de Normandie and Centre Universitaire de Recherche et D'Innovation en Biologie, Normandie University, UNIROUEN, INSERM, Rouen, France
| | - Cerra Maria Carmela
- Laboratory of Organ and System Physiology, Dept. of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| |
Collapse
|
34
|
Yamakawa N, Vanbeselaere J, Chang LY, Yu SY, Ducrocq L, Harduin-Lepers A, Kurata J, Aoki-Kinoshita KF, Sato C, Khoo KH, Kitajima K, Guerardel Y. Systems glycomics of adult zebrafish identifies organ-specific sialylation and glycosylation patterns. Nat Commun 2018; 9:4647. [PMID: 30405127 PMCID: PMC6220181 DOI: 10.1038/s41467-018-06950-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 09/26/2018] [Indexed: 12/31/2022] Open
Abstract
The emergence of zebrafish Danio rerio as a versatile model organism provides the unique opportunity to monitor the functions of glycosylation throughout vertebrate embryogenesis, providing insights into human diseases caused by glycosylation defects. Using a combination of chemical modifications, enzymatic digestion and mass spectrometry analyses, we establish here the precise glycomic profiles of eight individual zebrafish organs and demonstrate that the protein glycosylation and glycosphingolipid expression patterns exhibits exquisite specificity. Concomitant expression screening of a wide array of enzymes involved in the synthesis and transfer of sialic acids shows that the presence of organ-specific sialylation motifs correlates with the localized activity of the corresponding glycan biosynthesis pathways. These findings provide a basis for the rational design of zebrafish lines expressing desired glycosylation profiles.
Collapse
Affiliation(s)
- Nao Yamakawa
- Université de Lille, CNRS, UMR 8576 - UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000, Lille, France.,Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-8601, Japan
| | - Jorick Vanbeselaere
- Université de Lille, CNRS, UMR 8576 - UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000, Lille, France
| | - Lan-Yi Chang
- Université de Lille, CNRS, UMR 8576 - UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000, Lille, France.,Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
| | - Shin-Yi Yu
- Université de Lille, CNRS, UMR 8576 - UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000, Lille, France
| | - Lucie Ducrocq
- Université de Lille, CNRS, UMR 8576 - UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000, Lille, France
| | - Anne Harduin-Lepers
- Université de Lille, CNRS, UMR 8576 - UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000, Lille, France
| | - Junichi Kurata
- Faculty of Science and Engineering, Soka University, Hachioji, Tokyo, 192-8577, Japan
| | | | - Chihiro Sato
- Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-8601, Japan
| | - Kay-Hooi Khoo
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
| | - Ken Kitajima
- Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-8601, Japan
| | - Yann Guerardel
- Université de Lille, CNRS, UMR 8576 - UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000, Lille, France.
| |
Collapse
|
35
|
Sykes DJ, Suriyampola PS, Martins EP. Recent experience impacts social behavior in a novel context by adult zebrafish (Danio rerio). PLoS One 2018; 13:e0204994. [PMID: 30335773 PMCID: PMC6193632 DOI: 10.1371/journal.pone.0204994] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 09/18/2018] [Indexed: 12/12/2022] Open
Abstract
Many animals exhibit behavioral plasticity as they move between habitats seasonally, reside in fluctuating environments, or respond to human-induced environmental change. We know that physical environment during early development can have a lasting impact on behavior, and on the neural mechanisms that shape behavior. In adults, social context can have similar persistent effects on behavior and the brain. Here, we asked whether physical context impacts adult social behavior in a novel environment. We placed groups of adult zebrafish (Danio rerio) in two different physical contexts. After two weeks, we measured group behavior in a novel context, and found that zebrafish with recent experience in a more-complex physical environment charged each other more often and tended to form tighter shoals than did fish that had been housed in less-complex environments. These differences were present regardless of the novel context in which we assayed behavior, and were not easily explained by differences in activity level. Our results demonstrate the impact of recent experiences on adult behavior, and highlight the importance of physical as well as social history in predicting animal behavior in novel situations.
Collapse
Affiliation(s)
- Delawrence J. Sykes
- Department of Biology, Indiana University, Bloomington, Indiana, United States of America
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| | - Piyumika S. Suriyampola
- Department of Biology, Indiana University, Bloomington, Indiana, United States of America
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Emília P. Martins
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| |
Collapse
|
36
|
Qin H, Zhao A, Fu X. Chemical modulation of cell fates: in situ regeneration. SCIENCE CHINA-LIFE SCIENCES 2018; 61:1137-1150. [PMID: 30099708 DOI: 10.1007/s11427-018-9349-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 05/09/2018] [Indexed: 12/18/2022]
Abstract
Chemical modulation of cell fates has been widely used to promote tissue and organ regeneration. Small molecules can target the self-renewal, expansion, differentiation, and survival of endogenous stem cells for enhancing their regenerative power or induce dedifferentiation or transdifferentiation of mature cells into proliferative progenitors or specialized cell types needed for regeneration. Here, we discuss current progress and potential using small molecules to promote in vivo regenerative processes by regulating the cell fate. Current studies of small molecules in regeneration will provide insights into developing safe and efficient chemical approaches for in situ tissue repair and regeneration.
Collapse
Affiliation(s)
- Hua Qin
- Graduate School of Tianjin Medical University, Tianjin, 300070, China.,Cell Biology and Tissue Repair Laboratory, Key Laboratory of Wound Repair and Regeneration of PLA, the First Hospital Affiliated to the PLA General Hospital, Beijing, 100048, China
| | - Andong Zhao
- Graduate School of Tianjin Medical University, Tianjin, 300070, China.,Cell Biology and Tissue Repair Laboratory, Key Laboratory of Wound Repair and Regeneration of PLA, the First Hospital Affiliated to the PLA General Hospital, Beijing, 100048, China
| | - Xiaobing Fu
- Cell Biology and Tissue Repair Laboratory, Key Laboratory of Wound Repair and Regeneration of PLA, the First Hospital Affiliated to the PLA General Hospital, Beijing, 100048, China. .,College of Life Sciences, PLA General Hospital, PLA Medical College, Beijing, 100853, China.
| |
Collapse
|
37
|
Restoration of cardiac function after anaemia-induced heart failure in zebrafish. J Mol Cell Cardiol 2018; 121:223-232. [PMID: 30009777 DOI: 10.1016/j.yjmcc.2018.07.128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 06/28/2018] [Accepted: 07/11/2018] [Indexed: 01/22/2023]
Abstract
AIMS New therapeutic approaches are needed to fight against the growing epidemic of heart failure. Unlike mammals, zebrafish possess the incredible ability to regenerate cardiac tissue after acute trauma such as apical resection. Yet, the ability of zebrafish to recover after a chronic stress leading to heart failure has not been reported. The aim of this study was to test whether zebrafish can recover a normal cardiac function after anaemia-induced heart failure. METHODS AND RESULTS Eight- to ten-month-old zebrafish were treated with phenylhydrazine hydrochloride, an anaemia inducer, to generate heart failure. Treatment was stopped after 5 weeks and fish were followed-up for 3 weeks. Assessment of ventricular function by ultrasound at the end of the treatment revealed an increase in ventricle diameter (+47%) and a decrease in heart rate (-36%) and fractional shortening (-30%). A decrease in swim capacity was also observed (-31%). Tissue staining showed a thickening of the ventricular wall (5-fold), cell apoptosis and proliferation but no fibrosis. Expression of foetal genes, angiogenic factor and inflammation markers was increased, and β-adrenergic receptor-1 was decreased. Three weeks after phenylhydrazine hydrochloride withdrawal, all parameters returned to baseline and the fish recovered a normal cardiac function, tissue morphology and gene expression. CONCLUSIONS Zebrafish are able to completely recover from anaemia-induced heart failure. This model represents a unique opportunity to investigate the mechanisms of cardiac repair and may lead to the discovery of novel therapeutic targets of heart failure.
Collapse
|
38
|
Cardiac Stem Cells in the Postnatal Heart: Lessons from Development. Stem Cells Int 2018; 2018:1247857. [PMID: 30034478 PMCID: PMC6035836 DOI: 10.1155/2018/1247857] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 05/23/2018] [Indexed: 12/26/2022] Open
Abstract
Heart development in mammals is followed by a postnatal decline in cell proliferation and cell renewal from stem cell populations. A better understanding of the developmental changes in cardiac microenvironments occurring during heart maturation will be informative regarding the loss of adult regenerative potential. We reevaluate the adult heart's mitotic potential and the reported adult cardiac stem cell populations, as these are two topics of ongoing debate. The heart's early capacity for cell proliferation driven by progenitors and reciprocal signalling is demonstrated throughout development. The mature heart architecture and environment may be more restrictive on niches that can host progenitor cells. The engraftment issues observed in cardiac stem cell therapy trials using exogenous stem cells may indicate a lack of supporting stem cell niches, while tissue injury adds to a hostile microenvironment for transplanted cells. Engraftment may be improved by preconditioning the cultured stem cells and modulating the microenvironment to host these cells. These prospective areas of further research would benefit from a better understanding of cardiac progenitor interactions with their microenvironment throughout development and may lead to enhanced cardiac niche support for stem cell therapy engraftment.
Collapse
|
39
|
Marshall L, Girardot F, Demeneix BA, Coen L. Is adult cardiac regeneration absent in Xenopus laevis yet present in Xenopus tropicalis? Cell Biosci 2018; 8:31. [PMID: 29713454 PMCID: PMC5907698 DOI: 10.1186/s13578-018-0231-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/16/2018] [Indexed: 02/06/2023] Open
Abstract
We recently used an endoscopy-based resection method to explore the consequences of cardiac injury in adult Xenopus laevis, obtaining the result that the adult Xenopus heart is unable to regenerate. At 11 months post-amputation, cellular and biological marks of scarring persisted. We thus concluded that, contrary to urodeles and teleosts, adult anurans share a cardiac injury outcome similar to adult mammals. However, in their work published in this journal on the 13 December 2017, Liao et al. showed that the adult Xenopus tropicalis heart is capable of efficient, almost scar free regeneration, a result at odds with our previous observation. These findings contrast with and challenge the outcome of adult heart repair following injury in Xenopus species. Here we discuss the question of the intrinsic cardiac regenerative properties of an adult heart in anuran amphibians.
Collapse
Affiliation(s)
- Lindsey Marshall
- Evolution des Régulations Endocriniennes, Département Adaptation du vivant, UMR CNRS 7221, Muséum National d'Histoire Naturelle, Sorbonne Université, Paris, France
| | - Fabrice Girardot
- Evolution des Régulations Endocriniennes, Département Adaptation du vivant, UMR CNRS 7221, Muséum National d'Histoire Naturelle, Sorbonne Université, Paris, France
| | - Barbara A Demeneix
- Evolution des Régulations Endocriniennes, Département Adaptation du vivant, UMR CNRS 7221, Muséum National d'Histoire Naturelle, Sorbonne Université, Paris, France
| | - Laurent Coen
- Evolution des Régulations Endocriniennes, Département Adaptation du vivant, UMR CNRS 7221, Muséum National d'Histoire Naturelle, Sorbonne Université, Paris, France
| |
Collapse
|
40
|
Brown D, Samsa LA, Ito C, Ma H, Batres K, Arnaout R, Qian L, Liu J. Neuregulin-1 is essential for nerve plexus formation during cardiac maturation. J Cell Mol Med 2017; 22:2007-2017. [PMID: 29265764 PMCID: PMC5824398 DOI: 10.1111/jcmm.13408] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/30/2017] [Indexed: 01/08/2023] Open
Abstract
The Neuregulin‐1 (Nrg1)/ErbB pathway plays multiple, critical roles in early cardiac and nervous system development and has been implicated in both heart and nerve repair processes. However, the early embryonic lethality of mouse Nrg1 mutants precludes an analysis of Nrg1's function in later cardiac development and homeostasis. In this study, we generated a novel nrg1 null allele targeting all known isoforms of nrg1 in zebrafish and examined cardiac structural and functional parameters throughout development. We found that zebrafish nrg1 mutants instead survived until young adult stages when they exhibited reduced survivorship. This coincided with structural and functional defects in the developing juvenile and young adult hearts, as demonstrated by reduced intracardiac myocardial density, cardiomyocyte cell number, swimming performance and dysregulated heartbeat. Interestingly, nrg1 mutant hearts were missing long axons on the ventricle surface by standard length (SL) 5 mm, which preceded juvenile and adult cardiac defects. Given that the autonomic nervous system normally exerts fine control of cardiac output through this nerve plexus, these data suggest that Nrg1 may play a critical role in establishing the cardiac nerve plexus such that inadequate innervation leads to deficits in cardiac maturation, function and survival.
Collapse
Affiliation(s)
- Daniel Brown
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Leigh Ann Samsa
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - Cade Ito
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Hong Ma
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Karla Batres
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Rima Arnaout
- Department of Medicine, Division of Cardiology, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Li Qian
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Jiandong Liu
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|