999 results on '"heart regeneration"'
Search Results
602. Dry-contact microelectrode membranes for wireless detection of electrical phenotypes in neonatal mouse hearts
- Author
-
Zhao, Yu, Cao, Hung, Beebe, Tyler, Zhang, Hemin, Zhang, Xiaoxiao, Chang, Honglong, Scremin, Oscar, Lien, Ching-Ling, Tai, Yu-Chong, and Hsiai, Tzung K.
- Published
- 2015
- Full Text
- View/download PDF
603. Brain natriuretic peptide is able to stimulate cardiac progenitor cell proliferation and differentiation in murine hearts after birth
- Author
-
Bielmann, Christelle, Rignault-Clerc, Stéphanie, Liaudet, Lucas, Li, Feng, Kunieda, Tetsuo, Sogawa, Chizuru, Zehnder, Tamara, Waeber, Bernard, Feihl, François, and Rosenblatt-Velin, Nathalie
- Published
- 2014
- Full Text
- View/download PDF
604. Human ventricular unloading induces cardiomyocyte proliferation
- Author
-
Hesham A. Sadek, Sandeep R Das, Pradeep P.A. Mammen, Sonia Garg, Aroumougame Asaithamby, Shibani Mukherjee, Wataru Kimura, Diana C. Canseco, Salim Abdisalaam, and Souparno Bhattacharya
- Subjects
Male ,Cell cycle checkpoint ,heart regeneration ,DNA damage ,Heart Ventricles ,Mitosis ,Ataxia Telangiectasia Mutated Proteins ,DNA damage response ,DNA, Mitochondrial ,Mitochondria, Heart ,Histones ,medicine ,Aurora Kinase B ,Humans ,Myocytes, Cardiac ,Phosphorylation ,ventricular assist device ,Cell Proliferation ,Cell Size ,Cytokinesis ,Genetics ,Cell Nucleus ,heart failure ,Cell growth ,business.industry ,Middle Aged ,medicine.disease ,equipment and supplies ,Cell biology ,Cell nucleus ,medicine.anatomical_structure ,Heart failure ,Female ,Heart-Assist Devices ,Cardiology and Cardiovascular Medicine ,business ,Cardiomyopathies ,mechanical unloading - Abstract
Background The adult mammalian heart is incapable of meaningful regeneration after substantial cardiomyocyte loss, primarily due to the inability of adult cardiomyocytes to divide. Our group recently showed that mitochondria-mediated oxidative DNA damage is an important regulator of postnatal cardiomyocyte cell cycle arrest. However, it is not known whether mechanical load also plays a role in this process. We reasoned that the postnatal physiological increase in mechanical load contributes to the increase in mitochondrial content, with subsequent activation of DNA damage response (DDR) and permanent cell cycle arrest of cardiomyocytes. Objectives The purpose of this study was to test the effect of mechanical unloading on mitochondrial mass, DDR, and cardiomyocyte proliferation. Methods We examined the effect of human ventricular unloading after implantation of left ventricular assist devices (LVADs) on mitochondrial content, DDR, and cardiomyocyte proliferation in 10 matched left ventricular samples collected at the time of LVAD implantation (pre-LVAD) and at the time of explantation (post-LVAD). Results We found that post-LVAD hearts showed up to a 60% decrease in mitochondrial content and up to a 45% decrease in cardiomyocyte size compared with pre-LVAD hearts. Moreover, we quantified cardiomyocyte nuclear foci of phosphorylated ataxia telangiectasia mutated protein, an upstream regulator of the DDR pathway, and we found a significant decrease in the number of nuclear phosphorylated ataxia telangiectasia mutated foci in the post-LVAD hearts. Finally, we examined cardiomyocyte mitosis and cytokinesis and found a statistically significant increase in both phosphorylated histone H3–positive, and Aurora B–positive cardiomyocytes in the post-LVAD hearts. Importantly, these results were driven by statistical significance in hearts exposed to longer durations of mechanical unloading. Conclusions Prolonged mechanical unloading induces adult human cardiomyocyte proliferation, possibly through prevention of mitochondria-mediated activation of DDR.
- Published
- 2014
605. Cardiac tissue engineering: a reflection after a decade of hurry
- Author
-
Valentina eDi Felice, Rosario eBarone, Giorgia eNardone, Giancarlo eForte, Di Felice, V, Barone, R, Nardone, G, and Forte, G
- Subjects
Pathology ,medicine.medical_specialty ,heart regeneration ,Physiology ,cardiac progenitor cells ,Clinical uses of mesenchymal stem cells ,proto-tissues ,lcsh:Physiology ,Tissue engineering ,Physiology (medical) ,Medicine ,Induced pluripotent stem cell ,Stem cell transplantation for articular cartilage repair ,lcsh:QP1-981 ,business.industry ,Regeneration (biology) ,Mesenchymal stem cell ,Opinion Article ,tissue engineering ,scaffolds ,Stem cell ,business ,Neuroscience ,cardiac progenitor cells, proto-tissues, heart regeneration, tissue engineering, scaffolds, biomaterials ,biomaterials ,Adult stem cell - Abstract
The heart is a perfect machine whose mass is mainly composed of cardiomyocytes, but also fibroblasts, endothelial, smooth muscle, nervous, and immune cells are represented. One thousand million cardiomyocytes are estimated to be lost after myocardial infarction, their loss being responsible for the impairment in heart contractile function (Laflamme and Murry, 2005). The potential success of cardiac cell therapy relies almost completely on the ability of the implanted cells to differentiate toward mature cardiomyocytes. These cells must be able to reinforce the pumping activity of the injured heart in the absence of life-threatening arrhythmias due to electrophysiological incompatibility. These conditions can only be met if the newly formed cardiomyocytes can integrate electromechanically with the host tissue while getting appropriate vascularization from host- or donor-derived vessels. Together with the regeneration of the contractile component of the heart, attention must be paid to the search for novel methods to deliver appropriate vascularization to the ischemic areas and to the preservation of structures controlling the efficiency of blood pumping to the organism: the heart valves. Although fetal, neonatal, and adult cardiomyocytes have been tested for their actual ability to engraft diseased heart and improve its function (Dowell et al., 2003), most of the hopes in cardiac tissue regeneration relies on the possibility to use pluripotent and adult stem cells in targeted tissue engineering applications. In this respect, the set-up of protocols to obtain reprogrammed pluripotent stem cells (Takahashi and Yamanaka, 2006), a discovery awarded with the Nobel Prize in Physiology or Medicine in 2012, represents the demonstration that such advances in knowledge and technology will eventually lead to novel approaches to the treatment of incurable diseases. Unfortunately, notwithstanding a decade of intense investments, the results in terms of knowledge transferred to the clinical practice are very limited. The present issue addresses the topic of cardiac tissue regeneration from the viewpoint of stem cell biologists and tissue engineers, seeking for the most appropriate source of cells to replace dead myocardium, improve the organ structure and function, as well as exploring the most suitable delivery system for such cells. In the last couple of years a number of papers have been published on the ability of biomaterials and bio-constructs to drive the differentiation of stem cells for cardiac tissue engineering applications. Most of these studies used mesenchymal stem cells (MSCs) from bone marrow, adipose tissue or induced pluripotent stem cells (iPSCs). On the other hand, the contribution of cells derived from the heart tissues is here considered of outmost importance since these cells are believed to retain the potential to become highly differentiated cardiomyocytes.
- Published
- 2014
606. Network-based Approach Enabling Drug Repositioning for the Treatment of Myocardial Infarction
- Author
-
Androsova, Ganna, Fonds National de la Recherche - FnR [sponsor], Azuaje, Francisco [superviser], Schneider, Reinhard [superviser], Nazarov, Petr [member of the jury], Krause, Roland [member of the jury], and Centre de Recherche Public de la Santé - CRP SANTE [research center]
- Subjects
Multidisciplinaire, généralités & autres [F99] [Sciences du vivant] ,myocardial infarction ,heart regeneration ,WGCNA ,Multidisciplinary, general & others [F99] [Life sciences] ,zebrafish model ,cryoinjury - Abstract
Despite a notable reduction in incidence of acute myocardial infarction (MI), patients who experience it remain at risk for premature death and cardiac malfunction. The human cardiomyocytes are not able to achieve extensive regeneration upon MI. Remarkably, the adult zebrafish is able to achieve complete heart regeneration following amputation, cryoinjury or genetic ablation. This raises new potential opportunities on how to boost the heart healing capacity in humans. The objective of our research is to characterize the transcriptional network of the zebrafish heart regeneration, to describe underlying regulatory mechanisms, and to identify potential drugs capable to boost heart regeneration capacity. Having identified the gene co-expression patterns in the data from a zebrafish cryoinjury model, we constructed a weighted gene co-expression network. To detect candidate functional sub-networks (modules), we used two different network clustering approaches: a density-based (ClusterONE) and a topological overlap-based (Dynamic Hybrid) algorithms. We identified eighteen distinct modules associated with heart recovery upon cryoinjury. Functional enrichment analysis displayed that the modules are involved in different cellular processes crucial for heart regeneration, including: cell fate specification (p-value < 0.006) and migration (p-value < 0.047), cardiac cell differentiation (p-value < 3E-04), and various signaling events (p-value < 0.037). The visualization of the modules’ expression profiles confirmed the relevance of these functional enrichments. Among the candidate hub genes detected in the network, there are genes relevant to atherosclerosis treatment and inflammation during cardiac arrest. Among the top candidate drugs, there were drugs already reported to play therapeutic roles in heart disease, though the majority of the drugs have not been considered yet for myocardial infarction treatment. In conclusion, our findings provide insights into the complex regulatory mechanisms involved during heart regeneration in the zebrafish. These data will be useful for modeling specific network-based responses to heart injury, and for finding sensitive network points that may trigger or boost heart regeneration in the zebrafish, and possibly in mammals.
- Published
- 2014
607. ERBB2 triggers mammalian heart regeneration by promoting cardiomyocyte dedifferentiation and proliferation
- Author
-
Gabriele D'Uva, Yosef Yarden, Elad Bassat, Tal Konfino, Ori Brenner, Karen Weisinger, Julius Hegesh, Rachel Sarig, Jonathan Leor, Marina Lysenko, Michal Neeman, Mattia Lauriola, Richard P. Harvey, David Kain, Oren Yifa, Silvia Carvalho, Eldad Tzahor, Dana Rajchman, Yfat Yahalom-Ronen, Alla Aharonov, Gabriele D'Uva, Alla Aharonov, Mattia Lauriola, David Kain, Yfat Yahalom-Ronen, Silvia Carvalho, Karen Weisinger, Elad Bassat, Dana Rajchman, Oren Yifa, Marina Lysenko, Tal Konfino, Julius Hegesh, Ori Brenner, Michal Neeman, Yosef Yarden, Jonathan Leor, Rachel Sarig, Richard P. Harvey, and Eldad Tzahor
- Subjects
MAPK/ERK pathway ,cardiomyocyte hypertrophy ,Beta-catenin ,heart regeneration ,Time Factors ,Receptor, ErbB-2 ,Neuregulin-1 ,Myocardial Infarction ,regenerative medicine ,Editorials: Cell Cycle Features ,Time-Lapse Imaging ,Glycogen Synthase Kinase 3 ,GSK-3 ,cardiomyocyte dedifferentation ,Myocyte ,Animals ,Regeneration ,Myocytes, Cardiac ,ERBB2 ,Extracellular Signal-Regulated MAP Kinases ,Protein kinase B ,Cells, Cultured ,beta Catenin ,Cell Proliferation ,Mice, Knockout ,Glycogen Synthase Kinase 3 beta ,biology ,Dose-Response Relationship, Drug ,Regeneration (biology) ,Age Factors ,Cell Biology ,Cell Dedifferentiation ,Embryonic stem cell ,Magnetic Resonance Imaging ,Cell biology ,Disease Models, Animal ,Animals, Newborn ,cardiomyocyte proliferation ,biology.protein ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
The murine neonatal heart can regenerate after injury through cardiomyocyte (CM) proliferation, although this capacity markedly diminishes after the first week of life. Neuregulin-1 (NRG1) administration has been proposed as a strategy to promote cardiac regeneration. Here, using loss- and gain-of-function genetic tools, we explore the role of the NRG1 co-receptor ERBB2 in cardiac regeneration. NRG1-induced CM proliferation diminished one week after birth owing to a reduction in ERBB2 expression. CM-specific Erbb2 knockout revealed that ERBB2 is required for CM proliferation at embryonic/neonatal stages. Induction of a constitutively active ERBB2 (caERBB2) in neonatal, juvenile and adult CMs resulted in cardiomegaly, characterized by extensive CM hypertrophy, dedifferentiation and proliferation, differentially mediated by ERK, AKT and GSK3β/β-catenin signalling pathways. Transient induction of caERBB2 following myocardial infarction triggered CM dedifferentiation and proliferation followed by redifferentiation and regeneration. Thus, ERBB2 is both necessary for CM proliferation and sufficient to reactivate postnatal CM proliferative and regenerative potentials.
- Published
- 2014
608. Selecting biologically informative genes in co-expression networks with a centrality score
- Author
-
Francisco Azuaje
- Subjects
Microarrays ,Immunology ,Gene regulatory network ,Weighted networks ,RNA-Seq ,Context (language use) ,Computational biology ,Biology ,Gene co-expression networks ,General Biochemistry, Genetics and Molecular Biology ,Network hubs ,Animals ,Regeneration ,Gene Regulatory Networks ,Gene ,Ecology, Evolution, Behavior and Systematics ,Zebrafish ,Cancer ,Genetics ,Agricultural and Biological Sciences(all) ,Biochemistry, Genetics and Molecular Biology(all) ,Applied Mathematics ,Gene Expression Profiling ,Myocardium ,Research ,Centrality scores ,Heart ,Gene expression profiling ,Modeling and Simulation ,DNA microarray ,General Agricultural and Biological Sciences ,Centrality ,Biological network ,Heart regeneration - Abstract
Background Measures of node centrality in biological networks are useful to detect genes with critical functional roles. In gene co-expression networks, highly connected genes (i.e., candidate hubs) have been associated with key disease-related pathways. Although different approaches to estimating gene centrality are available, their potential biological relevance in gene co-expression networks deserves further investigation. Moreover, standard measures of gene centrality focus on binary interaction networks, which may not always be suitable in the context of co-expression networks. Here, I also investigate a method that identifies potential biologically meaningful genes based on a weighted connectivity score and indicators of statistical relevance. Results The method enables a characterization of the strength and diversity of co-expression associations in the network. It outperformed standard centrality measures by highlighting more biologically informative genes in different gene co-expression networks and biological research domains. As part of the illustration of the gene selection potential of this approach, I present an application case in zebrafish heart regeneration. The proposed technique predicted genes that are significantly implicated in cellular processes required for tissue regeneration after injury. Conclusions A method for selecting biologically informative genes from gene co-expression networks is provided, together with free open software. Reviewers This article was reviewed by Anthony Almudevar, Maciej M Kańduła (nominated by David P Kreil) and Christine Wells.
- Published
- 2014
609. Use of Echocardiography Reveals Reestablishment of Ventricular Pumping Efficiency and Partial Ventricular Wall Motion Recovery upon Ventricular Cryoinjury in the Zebrafish
- Author
-
Luis Jesús Jiménez-Borreguero, Juan Manuel González-Rosa, Gabriela Guzmán-Martínez, Ines J. Marques, Héctor Sánchez-Iranzo, Nadia Mercader, European Commission, Fundación ProCNIC, Ministerio de Economía y Competitividad (España), Comunidad de Madrid, European Research Council, and UAM. Departamento de Medicina
- Subjects
Pathology ,Muscle Physiology ,CARDIAC REGENERATION ,Muscle Functions ,Physiology ,Myocardial Infarction ,lcsh:Medicine ,ADULT ZEBRAFISH ,Cardiovascular Physiology ,DISEASE ,Heart Regeneration ,REPRODUCIBILITY ,Morphogenesis ,Ventricular Dysfunction ,Biomechanics ,Myocardial infarction ,lcsh:Science ,Zebrafish ,Multidisciplinary ,Cardiac cycle ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,3. Good health ,Cardiovascular physiology ,Cold Temperature ,medicine.anatomical_structure ,Echocardiography ,Cardiology ,cardiovascular system ,2D-echocardiography ,Research Article ,Muscle Contraction ,HEART REGENERATION ,Cardiac function curve ,EXPRESSION ,medicine.medical_specialty ,Medicina ,Heart Ventricles ,610 Medicine & health ,Real-Time Polymerase Chain Reaction ,Internal medicine ,medicine ,Regeneration ,Animals ,RNA, Messenger ,business.industry ,Regeneration (biology) ,lcsh:R ,Ventricular ,Biology and Life Sciences ,Cardiac Ventricle ,medicine.disease ,biology.organism_classification ,Fibrosis ,GENE ,MYOCARDIAL-INFARCTION ,Ventricle ,lcsh:Q ,business ,Organism Development ,Developmental Biology - Abstract
The authors confirm that all data underlying the findings are fully available without restriction. All relevant data are within the paper and its Supporting Information files, Aims: While zebrafish embryos are amenable to in vivo imaging, allowing the study of morphogenetic processes during development, intravital imaging of adults is hampered by their small size and loss of transparency. The use of adult zebrafish as a vertebrate model of cardiac disease and regeneration is increasing at high speed. It is therefore of great importance to establish appropriate and robust methods to measure cardiac function parameters. Methods and Results: Here we describe the use of 2D-echocardiography to study the fractional volume shortening and segmental wall motion of the ventricle. Our data show that 2D-echocardiography can be used to evaluate cardiac injury and also to study recovery of cardiac function. Interestingly, our results show that while global systolic function recovered following cardiac cryoinjury, ventricular wall motion was only partially restored. Conclusion: Cryoinjury leads to long-lasting impairment of cardiac contraction, partially mimicking the consequences of myocardial infarction in humans. Functional assessment of heart regeneration by echocardiography allows a deeper understanding of the mechanisms of cardiac regeneration and has the advantage of being easily transferable to other cardiovascular zebrafish disease models, Funding was from the Fundación CNIC Carlos III, the Fundación ProCNIC, the Spanish Ministry of Economy and Competitiveness (Tercel and BFU2011-25297 to N.M., FPU AP2008-00546 to J.M.G.-R. and FPU12/03007 to H.S-I.), the Community of Madrid (FIBROTEAM S2010/BMD- 2321 to N.M), PIEF-GA-2012-330728 to I.J.M. and an ERC Starting grant 337703 – zebraHeart to N.M.
- Published
- 2014
610. Targeting pleiotropic signaling pathways to control adult cardiac stem cell fate and function
- Author
-
Gabriele Grassi, Giancarlo Forte, Jakub Jelinek, Stefania Pagliari, S., Pagliari, J., Jelinek, Grassi, Gabriele, and Forte, Giancarlo
- Subjects
Cell signaling ,Pathology ,medicine.medical_specialty ,heart regeneration ,Physiology ,Review Article ,Biology ,lcsh:Physiology ,stem cell homeostasis ,Physiology (medical) ,cardiacstemcell ,medicine ,cell signaling ,Progenitor cell ,Tissue homeostasis ,cellsignaling ,lcsh:QP1-981 ,Regeneration (biology) ,differentiation ,stemcellhomeostasis ,Cell biology ,heartregeneration ,Signal transduction ,Stem cell ,cardiac stem cell ,Function (biology) ,Homeostasis - Abstract
The identification of different pools of cardiac progenitor cells resident in the adult mammalian heart opened a new era in heart regeneration as a means to restore the loss of functional cardiac tissue and overcome the limited availability of donor organs. Indeed, resident stem cells are believed to participate to tissue homeostasis and renewal in healthy and damaged myocardium although their actual contribution to these processes remain unclear. The poor outcome in terms of cardiac regeneration following tissue damage point out at the need for a deeper understanding of the molecular mechanisms controlling CPC behavior and fate determination before new therapeutic strategies can be developed. The regulation of cardiac resident stem cell fate and function is likely to result from the interplay between pleiotropic signaling pathways as well as tissue- and cell-specific regulators. Such a modular interaction – which has already been described in the nucleus of a number of different cells where transcriptional complexes form to activate specific gene programs - would account for the unique responses of cardiac progenitors to general and tissue-specific stimuli.The study of the molecular determinants involved in cardiac stem/progenitor cell regulatory mechanisms may shed light on the processes of cardiac homeostasis in health and disease and thus provide clues on the actual feasibility of cardiac cell therapy through tissue-specific progenitors.
- Published
- 2014
611. A Clonal Analysis of Zebrafish Heart Morphogenesis and Regeneration
- Author
-
Gupta, Vikas and Poss, Kenneth D
- Subjects
Heart Regeneration ,Organogenesis ,Developmental biology ,Clonal Analysis ,Zebrafish - Abstract
As vertebrate embryos grow and develop into adults, their organs must acquire mass and mature tissue architecture to maintain proper homeostasis. While juvenile growth encompasses a significant portion of life, relatively little is known about how individual cells proliferate, with respect to one another, to orchestrate this final maturation. For its simplicity and ease of genetic manipulations, the teleost zebrafish (Danio rerio) was used to understand how the proliferative outputs of individual cells generate an organ from embryogenesis into adulthood. To define the proliferative outputs of individual cells, a multicolor clonal labeling approach was taken that visualized a large number of cardiomyocyte clones within the zebrafish heart. This Brainbow technique utilizes Cre-loxP mediated recombination to assign cells upwards of ~90 unique genetic tags. These tags are comprised of the differential expression of 3 fluorescent proteins, which combine to give rise to spectrally distinct colors that represent these genetic tags. Tagging of individual cardiomyocytes was induced early in development, when the wall of the cardiac ventricle is a single myocyte thick. Single cell cardiomyocyte clones within this layer expanded laterally in a developmentally plastic manner into patches of variable shapes and sizes as animals grew into juveniles. As maturation continued into adulthood, a new lineage of cortical muscle appeared at the base of the ventricle and enveloped the ventricle in a wave of proliferation that fortified the wall to make it several myocytes thick. This outer cortical layer was formed from a small number (~8) of dominant cortical myocyte clones that originated from trabecular myocytes. These trabecular myocytes were found to gain access to the ventricular surface through rare breaches within the single cell thick ventricular wall, before proliferating over the surface of the ventricle. These results demonstrated an unappreciated dynamic juvenile remodeling event that generated the adult ventricular wall. During adult zebrafish heart regeneration, the primary source of regenerating cardiomyocytes stems from this outer wall of muscle. Regenerating cardiomyocytes within this outer layer of muscle are specifically marked by the cardiac transcription factor gene gata4, which they continue to express as they proliferate into the wound area.Using heart regeneration to guide investigation of juvenile cortical layer formation, we found that both processes shared similar molecular and tissue specific responses including expression and requirement of gata4. Additional markers suggested that juvenile hearts were under stress and that this stress could play a role to initiate cortical morphogenesis. Indeed, experimental injury or a physiologic increase in stress to juvenile hearts caused the ectopic appearance of cortical muscle, demonstrating that injury could trigger premature morphogenesis.These studies detail the cardiomyocyte proliferative events that shape the heart and identify molecular parallels that exist between regeneration and cortical layer formation. They show that adult zebrafish heart regeneration utilizes an injury/stress responsive program that was first used to remodel the heart during juvenile growth.
- Published
- 2014
612. Zebrafish as a model of cardiac disease
- Author
-
Wilkinson, R. N., Jopling, Chris, Eeden, F. J., Institut de Génomique Fonctionnelle (IGF), and Université de Montpellier (UM)-Université Montpellier 1 (UM1)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Montpellier 2 - Sciences et Techniques (UM2)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Regeneration/physiology ,Animal ,Cardiomyopathy ,Disease model ,[SDV]Life Sciences [q-bio] ,Heart development ,Electrophysiological Phenomena ,Cardiac disease ,Disease Models ,Heart/embryology/physiology ,Congenital heart defects ,Animals ,Heart Diseases/*pathology ,Heart regeneration ,Zebrafish ,Arrhythmia ,Zebrafish/embryology/*physiology - Abstract
International audience; The zebrafish has been rapidly adopted as a model for cardiac development and disease. The transparency of the embryo, its limited requirement for active oxygen delivery, and ease of use in genetic manipulations and chemical exposure have made it a powerful alternative to rodents. Novel technologies like TALEN/CRISPR-mediated genome engineering and advanced imaging methods will only accelerate its use. Here, we give an overview of heart development and function in the fish and highlight a number of areas where it is most actively contributing to the understanding of cardiac development and disease. We also review the current state of research on a feature that we only could wish to be conserved between fish and human; cardiac regeneration.
- Published
- 2014
613. Heart regeneration, stem cells, and cytokines
- Author
-
Lixin Jia, Chuan Wang, Jie Du, and Na Li
- Subjects
business.industry ,Regeneration (biology) ,Mesenchymal stem cell ,lcsh:R ,lcsh:Medicine ,Review ,General Medicine ,Stem cells ,Neovascularization ,Paracrine signalling ,Immunology ,microRNA ,Cancer research ,Medicine ,Cytokines ,Cytokine secretion ,Paracrine mechanisms ,medicine.symptom ,Signal transduction ,Stem cell ,business ,Heart regeneration - Abstract
The human heart has limited regenerative capacity, which makes the reparative response after the cardiac infarction quite challenging. During the last decade, stem cells have become promising candidates for heart repair, owing to their potent differentiation capacity and paracrine cytokine secretion. Among the different types of stem cells, mesenchymal stem cells have high proliferative potential and secrete numerous cytokines, growth factors, and microRNAs. The paracrine cytokines play important roles in cardiac regeneration, neovascularization, anti-apoptosis, and anti-remodeling mechanisms, among others. This review summarizes the cytokines secreted by stem cells and their relative signaling pathways, which represent key mechanisms for heart regeneration and may serve as a promising future therapeutic strategy for myocardial infarction patients.
- Published
- 2014
614. Transcriptional response to cardiac injury in the zebrafish: systematic identification of genes with highly concordant activity across in vivo models
- Author
-
Céline Jeanty, Sophie Rodius, Petr V. Nazarov, Francisco Azuaje, Juan Manuel González-Rosa, Mark Ibberson, Isabel A. Nepomuceno-Chamorro, Ioannis Xenarios, Nadia Mercader, Ricardo Costa, Luxembourg National Research Fund, Swiss National Research Foundation, INFUSED project, Ministerio de Economía y Competitividad (España), Comunidad de Madrid, Universidad de Sevilla. Departamento de Lenguajes y Sistemas Informáticos, Universidad de Sevilla. TIC134: Sistemas Informáticos, Ministerio de Economía y Competitividad (MINECO). España, and Comunidad Autónoma de Madrid
- Subjects
Transcriptional association networks ,EXPRESSION ,Heart Injury ,Time Factors ,Ventricular cryoinjury ,INDUCED MYOCARDIAL-INFARCTION ,Microarray ,610 Medicine & health ,Computational biology ,Biology ,Periostin ,Real-Time Polymerase Chain Reaction ,Bioinformatics ,Transcriptome ,Cytochrome P-450 Enzyme System ,Ventricular amputation ,In vivo ,Endopeptidases ,Genetics ,Animals ,Regeneration ,PERIOSTIN ,INTERACTION NETWORKS ,Zebrafish ,Oligonucleotide Array Sequence Analysis ,HYPERTROPHIC CARDIOMYOPATHY ,Microarray analysis techniques ,Myocardium ,Myocardial infarction ,Heart regeneration ,Transcriptional responses ,PROLIFERATION ,Computational Biology ,Heart ,NEMALINE MYOPATHY ,Zebrafish Proteins ,CARDIOVASCULAR-SYSTEM ,biology.organism_classification ,Disease Models, Animal ,Heart Injuries ,Tumor Suppressor Protein p53 ,DNA microarray ,STEM-CELLS ,Research Article ,Biotechnology - Abstract
Background: Zebrafish is a clinically-relevant model of heart regeneration. Unlike mammals, it has a remarkable heart repair capacity after injury, and promises novel translational applications. Amputation and cryoinjury models are key research tools for understanding injury response and regeneration in vivo. An understanding of the transcriptional responses following injury is needed to identify key players of heart tissue repair, as well as potential targets for boosting this property in humans. Results: We investigated amputation and cryoinjury in vivo models of heart damage in the zebrafish through unbiased, integrative analyses of independent molecular datasets. To detect genes with potential biological roles, we derived computational prediction models with microarray data from heart amputation experiments. We focused on a top-ranked set of genes highly activated in the early post-injury stage, whose activity was further verified in independent microarray datasets. Next, we performed independent validations of expression responses with qPCR in a cryoinjury model. Across in vivo models, the top candidates showed highly concordant responses at 1 and 3 days post-injury, which highlights the predictive power of our analysis strategies and the possible biological relevance of these genes. Top candidates are significantly involved in cell fate specification and differentiation, and include heart failure markers such as periostin, as well as potential new targets for heart regeneration. For example, ptgis and ca2 were overexpressed, while usp2a, a regulator of the p53 pathway, was down-regulated in our in vivo models. Interestingly, a high activity of ptgis and ca2 has been previously observed in failing hearts from rats and humans. Conclusions: We identified genes with potential critical roles in the response to cardiac damage in the zebrafish. Their transcriptional activities are reproducible in different in vivo models of cardiac injury. This research was supported by the INTER program of Luxembourg's National Research Fund (FNR) and the Swiss National Research Foundation (SNF), INFUSED project (www.infused-project.eu). NM acknowledges support from Spanish Ministry of Economy and Competivity (BFU2011-25297 and TerCel projects) and the Comunidad de Madrid (P2010/BMD-2321). Sí
- Published
- 2014
615. Network-based Approach Enabling Drug Repositioning for the Treatment of Myocardial Infarction
- Author
-
Centre de Recherche Public de la Santé - CRP SANTE [research center], Fonds National de la Recherche - FnR [sponsor], Androsova, Ganna, Centre de Recherche Public de la Santé - CRP SANTE [research center], Fonds National de la Recherche - FnR [sponsor], and Androsova, Ganna
- Abstract
Despite a notable reduction in incidence of acute myocardial infarction (MI), patients who experience it remain at risk for premature death and cardiac malfunction. The human cardiomyocytes are not able to achieve extensive regeneration upon MI. Remarkably, the adult zebrafish is able to achieve complete heart regeneration following amputation, cryoinjury or genetic ablation. This raises new potential opportunities on how to boost the heart healing capacity in humans. The objective of our research is to characterize the transcriptional network of the zebrafish heart regeneration, to describe underlying regulatory mechanisms, and to identify potential drugs capable to boost heart regeneration capacity. Having identified the gene co-expression patterns in the data from a zebrafish cryoinjury model, we constructed a weighted gene co-expression network. To detect candidate functional sub-networks (modules), we used two different network clustering approaches: a density-based (ClusterONE) and a topological overlap-based (Dynamic Hybrid) algorithms. We identified eighteen distinct modules associated with heart recovery upon cryoinjury. Functional enrichment analysis displayed that the modules are involved in different cellular processes crucial for heart regeneration, including: cell fate specification (p-value < 0.006) and migration (p-value < 0.047), cardiac cell differentiation (p-value < 3E-04), and various signaling events (p-value < 0.037). The visualization of the modules’ expression profiles confirmed the relevance of these functional enrichments. Among the candidate hub genes detected in the network, there are genes relevant to atherosclerosis treatment and inflammation during cardiac arrest. Among the top candidate drugs, there were drugs already reported to play therapeutic roles in heart disease, though the majority of the drugs have not been considered yet for myocardial infarction treatment. In conclusion, our findings provide insights into the complex regul
- Published
- 2014
616. Transcriptional response to cardiac injury in the zebrafish: systematic identification of genes with highly concordant activity across in vivo models
- Author
-
Universidad de Sevilla. Departamento de Lenguajes y Sistemas Informáticos, Universidad de Sevilla. TIC134: Sistemas Informáticos, Ministerio de Economía y Competitividad (MINECO). España, Comunidad Autónoma de Madrid, Rodius, Sophie, Nazarov, Petr V., Nepomuceno Chamorro, Isabel de los Ángeles, Jeanty, Céline, González Rosa, Juan Manuel, Ibberson, Mark, Benites da Costa, Ricardo M., Xenarios, Ioannis, Mercader, Nadia, Azuaje, Francisco, Universidad de Sevilla. Departamento de Lenguajes y Sistemas Informáticos, Universidad de Sevilla. TIC134: Sistemas Informáticos, Ministerio de Economía y Competitividad (MINECO). España, Comunidad Autónoma de Madrid, Rodius, Sophie, Nazarov, Petr V., Nepomuceno Chamorro, Isabel de los Ángeles, Jeanty, Céline, González Rosa, Juan Manuel, Ibberson, Mark, Benites da Costa, Ricardo M., Xenarios, Ioannis, Mercader, Nadia, and Azuaje, Francisco
- Abstract
Background: Zebrafish is a clinically-relevant model of heart regeneration. Unlike mammals, it has a remarkable heart repair capacity after injury, and promises novel translational applications. Amputation and cryoinjury models are key research tools for understanding injury response and regeneration in vivo. An understanding of the transcriptional responses following injury is needed to identify key players of heart tissue repair, as well as potential targets for boosting this property in humans. Results: We investigated amputation and cryoinjury in vivo models of heart damage in the zebrafish through unbiased, integrative analyses of independent molecular datasets. To detect genes with potential biological roles, we derived computational prediction models with microarray data from heart amputation experiments. We focused on a top-ranked set of genes highly activated in the early post-injury stage, whose activity was further verified in independent microarray datasets. Next, we performed independent validations of expression responses with qPCR in a cryoinjury model. Across in vivo models, the top candidates showed highly concordant responses at 1 and 3 days post-injury, which highlights the predictive power of our analysis strategies and the possible biological relevance of these genes. Top candidates are significantly involved in cell fate specification and differentiation, and include heart failure markers such as periostin, as well as potential new targets for heart regeneration. For example, ptgis and ca2 were overexpressed, while usp2a, a regulator of the p53 pathway, was down-regulated in our in vivo models. Interestingly, a high activity of ptgis and ca2 has been previously observed in failing hearts from rats and humans. Conclusions: We identified genes with potential critical roles in the response to cardiac damage in the zebrafish. Their transcriptional activities are reproducible in different in vivo models of cardiac injury
- Published
- 2014
617. Coronary Revascularization During Heart Regeneration Is Regulated by Epicardial and Endocardial Cues and Forms a Scaffold for Cardiomyocyte Repopulation.
- Author
-
Marín-Juez R, El-Sammak H, Helker CSM, Kamezaki A, Mullapuli ST, Bibli SI, Foglia MJ, Fleming I, Poss KD, and Stainier DYR
- Subjects
- Animals, Cell Proliferation physiology, Chemokine CXCL12 metabolism, Cues, Endocardium physiology, Heart physiology, Heart Ventricles metabolism, Myocardial Revascularization methods, Myocytes, Cardiac metabolism, Pericardium physiology, Receptors, CXCR4 metabolism, Signal Transduction physiology, Wound Healing physiology, Zebrafish metabolism, Zebrafish Proteins metabolism, Myocytes, Cardiac physiology, Neovascularization, Physiologic physiology, Regeneration physiology
- Abstract
Defective coronary network function and insufficient blood supply are both cause and consequence of myocardial infarction. Efficient revascularization after infarction is essential to support tissue repair and function. Zebrafish hearts exhibit a remarkable ability to regenerate, and coronary revascularization initiates within hours of injury, but how this process is regulated remains unknown. Here, we show that revascularization requires a coordinated multi-tissue response culminating with the formation of a complex vascular network available as a scaffold for cardiomyocyte repopulation. During a process we term "coronary-endocardial anchoring," new coronaries respond by sprouting (1) superficially within the regenerating epicardium and (2) intra-ventricularly toward the activated endocardium. Mechanistically, superficial revascularization is guided by epicardial Cxcl12-Cxcr4 signaling and intra-ventricular sprouting by endocardial Vegfa signaling. Our findings indicate that the injury-activated epicardium and endocardium support cardiomyocyte replenishment initially through the guidance of coronary sprouting. Simulating this process in the injured mammalian heart should help its healing., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
618. Prolonged neutrophil retention in the wound impairs zebrafish heart regeneration after cryoinjury.
- Author
-
Xu S, Xie F, Tian L, Manno SH, Manno FAM 3rd, and Cheng SH
- Subjects
- Animals, Cryopreservation veterinary, Heart Injuries etiology, Heart Injuries physiopathology, Neutrophils immunology, Freezing adverse effects, Heart physiology, Regeneration, Signal Transduction physiology, Zebrafish physiology
- Abstract
Neutrophils are the first line defenders in the innate immune response, and rapidly migrate to an infected or injured area. Recently, bidirectional migration of neutrophils to the wound and the corresponding functions have become popular research pursuits. In zebrafish larvae, CXCR1/CXCL8 is the predominant chemoattractant pathway to recruit neutrophil to wound, while CXCR2/CXCL8 pathway mediate neutrophil dispersal in wound after injury. Here, we found that both CXCR1/CXCL8 and LTB4/BLT1 signals are activated in zebrafish heart after cryoinjury. And with a CXCR1/2 selective inhibitor (SB225002) treatment, the recruitment of neutrophils was not affected, but reverse migration of neutrophils was inhibited after cryoinjury of heart. We suggested that the neutrophil recruitment to cryoinjured area might be mediated by LTB4/BLT1 signals at the presence of SB225002. Therefore, SB225002 treatment resulted more accumulation and long retention of neutrophils in the injured heart. The long retention of neutrophils in the wound promoted revascularization in the injured heart; however, the AKT/mTOR pathway was inhibited and the regeneration was impaired. Our findings suggest that retention of neutrophils is a well-orchestrated process and might regulate regeneration by the AKT/mTOR pathway., (Copyright © 2019. Published by Elsevier Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
619. Recent advances in myocardial regeneration strategy.
- Author
-
Sheng K, Nie Y, and Gao B
- Subjects
- Animals, Humans, Cell Proliferation, Heart physiology, Heart Diseases therapy, Myocytes, Cardiac cytology, Regeneration
- Published
- 2019
- Full Text
- View/download PDF
620. Adult sox10 + Cardiomyocytes Contribute to Myocardial Regeneration in the Zebrafish.
- Author
-
Sande-Melón M, Marques IJ, Galardi-Castilla M, Langa X, Pérez-López M, Botos MA, Sánchez-Iranzo H, Guzmán-Martínez G, Ferreira Francisco DM, Pavlinic D, Benes V, Bruggmann R, and Mercader N
- Subjects
- Animals, Cell Proliferation, Cells, Cultured, Heart physiology, Myocytes, Cardiac physiology, SOXE Transcription Factors genetics, Zebrafish, Zebrafish Proteins genetics, Myocytes, Cardiac metabolism, Regeneration, SOXE Transcription Factors metabolism, Zebrafish Proteins metabolism
- Abstract
During heart regeneration in the zebrafish, fibrotic tissue is replaced by newly formed cardiomyocytes derived from preexisting ones. It is unclear whether the heart is composed of several cardiomyocyte populations bearing different capacity to replace lost myocardium. Here, using sox10 genetic fate mapping, we identify a subset of preexistent cardiomyocytes in the adult zebrafish heart with a distinct gene expression profile that expanded after cryoinjury. Genetic ablation of sox10
+ cardiomyocytes impairs cardiac regeneration, revealing that these cells play a role in heart regeneration., (Copyright © 2019 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
621. H3K27me3-mediated silencing of structural genes is required for zebrafish heart regeneration.
- Author
-
Ben-Yair R, Butty VL, Busby M, Qiu Y, Levine SS, Goren A, Boyer LA, Burns CG, and Burns CE
- Subjects
- Animals, Cell Proliferation, Cytokinesis, Cytoskeleton metabolism, Gene Expression Regulation, Developmental, Methylation, Myocytes, Cardiac cytology, Myocytes, Cardiac metabolism, Sarcomeres metabolism, Gene Silencing, Heart physiology, Histones metabolism, Lysine metabolism, Regeneration physiology, Zebrafish genetics, Zebrafish physiology
- Abstract
Deciphering the genetic and epigenetic regulation of cardiomyocyte proliferation in organisms that are capable of robust cardiac renewal, such as zebrafish, represents an attractive inroad towards regenerating the human heart. Using integrated high-throughput transcriptional and chromatin analyses, we have identified a strong association between H3K27me3 deposition and reduced sarcomere and cytoskeletal gene expression in proliferative cardiomyocytes following cardiac injury in zebrafish. To move beyond an association, we generated an inducible transgenic strain expressing a mutant version of histone 3, H3.3
K27M , that inhibits H3K27me3 catalysis in cardiomyocytes during the regenerative window. Hearts comprising H3.3K27M -expressing cardiomyocytes fail to regenerate, with wound edge cells showing heightened expression of structural genes and prominent sarcomeres. Although cell cycle re-entry was unperturbed, cytokinesis and wound invasion were significantly compromised. Collectively, our study identifies H3K27me3-mediated silencing of structural genes as requisite for zebrafish heart regeneration and suggests that repression of similar structural components in the border zone of an infarcted human heart might improve its regenerative capacity., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2019. Published by The Company of Biologists Ltd.)- Published
- 2019
- Full Text
- View/download PDF
622. Proteomics Analysis of Extracellular Matrix Remodeling During Zebrafish Heart Regeneration.
- Author
-
Garcia-Puig A, Mosquera JL, Jiménez-Delgado S, García-Pastor C, Jorba I, Navajas D, Canals F, and Raya A
- Subjects
- Animals, Biomechanical Phenomena, Extracellular Matrix ultrastructure, Extracellular Matrix Proteins analysis, Extracellular Matrix Proteins metabolism, Microscopy, Atomic Force, Proteomics methods, Zebrafish, Zebrafish Proteins genetics, Zebrafish Proteins metabolism, Extracellular Matrix physiology, Heart physiology, Myocardium cytology, Regeneration physiology, Zebrafish Proteins analysis
- Abstract
Adult zebrafish, in contrast to mammals, are able to regenerate their hearts in response to injury or experimental amputation. Our understanding of the cellular and molecular bases that underlie this process, although fragmentary, has increased significantly over the last years. However, the role of the extracellular matrix (ECM) during zebrafish heart regeneration has been comparatively rarely explored. Here, we set out to characterize the ECM protein composition in adult zebrafish hearts, and whether it changed during the regenerative response. For this purpose, we first established a decellularization protocol of adult zebrafish ventricles that significantly enriched the yield of ECM proteins. We then performed proteomic analyses of decellularized control hearts and at different times of regeneration. Our results show a dynamic change in ECM protein composition, most evident at the earliest (7 days postamputation) time point analyzed. Regeneration associated with sharp increases in specific ECM proteins, and with an overall decrease in collagens and cytoskeletal proteins. We finally tested by atomic force microscopy that the changes in ECM composition translated to decreased ECM stiffness. Our cumulative results identify changes in the protein composition and mechanical properties of the zebrafish heart ECM during regeneration., (© 2019 Garcia-Puig et al.)
- Published
- 2019
- Full Text
- View/download PDF
623. Opioid receptors and opioid peptides in the cardiomyogenesis of mouse embryonic stem cells.
- Author
-
Šínová R, Kudová J, Nešporová K, Karel S, Šuláková R, Velebný V, and Kubala L
- Subjects
- Animals, Cell Differentiation physiology, Mice, Mouse Embryonic Stem Cells metabolism, Myocytes, Cardiac physiology, Regeneration physiology, Mouse Embryonic Stem Cells cytology, Myocardium cytology, Myocytes, Cardiac cytology, Opioid Peptides metabolism, Receptors, Opioid metabolism
- Abstract
The stimulation of myocardium repair is restricted due to the limited understanding of heart regeneration. Interestingly, endogenous opioid peptides such as dynorphins and enkephalins are suggested to support this process. However, the mechanism-whether through the stimulation of the regenerative capacity of cardiac stem cells or through effects on other cell types in the heart-is still not completely understood. Thus, a model of the spontaneous cardiomyogenic differentiation of mouse embryonic stem (mES) cells via the formation of embryoid bodies was used to describe changes in the expression and localization of opioid receptors within cells during the differentiation process and the potential of the selected opioid peptides, dynorphin A and B, and methionin-enkephalins and leucin-enkephalins, to modulate cardiomyogenic differentiation in vitro. The expressions of both κ- and δ-opioid receptors significantly increased during mES cell differentiation. Moreover, their primary colocalization with the nucleus was followed by their growing presence on the cytoplasmic membrane with increasing mES cell differentiation status. Interestingly, dynorphin B enhanced the downregulation gene expression of Oct4 characteristic of the pluripotent phenotype. Further, dynorphin B also increased cardiomyocyte-specific Nkx2.5 gene expression. However, neither dynorphin A nor methionin-enkephalins and leucin-enkephalins exhibited any significant effects on the course of mES cell differentiation. In conclusion, despite the increased expression of opioid receptors and some enhancement of mES cell differentiation by dynorphin B, the overall data do not support the notion that opioid peptides have a significant potential to promote the spontaneous cardiomyogenesis of mES cells in vitro., (© 2018 Wiley Periodicals, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
624. Cardiac Reprogramming Factors Synergistically Activate Genome-wide Cardiogenic Stage-Specific Enhancers.
- Author
-
Hashimoto H, Wang Z, Garry GA, Malladi VS, Botten GA, Ye W, Zhou H, Osterwalder M, Dickel DE, Visel A, Liu N, Bassel-Duby R, and Olson EN
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors metabolism, Cells, Cultured, Cellular Reprogramming, ErbB Receptors genetics, GATA4 Transcription Factor genetics, Gene Regulatory Networks, Genome-Wide Association Study, MEF2 Transcription Factors genetics, Mice, Mice, Inbred C57BL, Signal Transduction, T-Box Domain Proteins genetics, ErbB Receptors metabolism, Fibroblasts physiology, Myocytes, Cardiac physiology
- Abstract
The cardiogenic transcription factors (TFs) Mef2c, Gata4, and Tbx5 can directly reprogram fibroblasts to induced cardiac-like myocytes (iCLMs), presenting a potential source of cells for cardiac repair. While activity of these TFs is enhanced by Hand2 and Akt1, their genomic targets and interactions during reprogramming are not well studied. We performed genome-wide analyses of cardiogenic TF binding and enhancer profiling during cardiac reprogramming. We found that these TFs synergistically activate enhancers highlighted by Mef2c binding sites and that Hand2 and Akt1 coordinately recruit other TFs to enhancer elements. Intriguingly, these enhancer landscapes collectively resemble patterns of enhancer activation during embryonic cardiogenesis. We further constructed a cardiac reprogramming gene regulatory network and found repression of EGFR signaling pathway genes. Consistently, chemical inhibition of EGFR signaling augmented reprogramming. Thus, by defining epigenetic landscapes these findings reveal synergistic transcriptional activation across a broad landscape of cardiac enhancers and key signaling pathways that govern iCLM reprogramming., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
625. Rat Left Ventricular Cardiomyocytes Characterization in the Process of Postinfarction Myocardial Remodeling.
- Author
-
Baidyuk EV, Sakuta GA, Vorobev ML, Stepanov AV, Karpov AA, Rogoza OV, and Kudryavtsev BN
- Subjects
- Animals, Cell Proliferation genetics, Hypertrophy, Male, Myocardium ultrastructure, Myocytes, Cardiac ultrastructure, Ploidies, Rats, Rats, Wistar, Regeneration genetics, Sarcomeres pathology, Time Factors, Myocardial Infarction pathology, Myocardium cytology, Myocardium pathology, Myocytes, Cardiac cytology, Myocytes, Cardiac pathology, Sarcomeres ultrastructure
- Abstract
Ischemic lesions of the heart, including myocardial infarction, are the most common pathologies of human cardiovascular system. Despite all the research and achievements of medicine in this field, the mortality from this disease remains heavy. Therefore, studying of processes occurring in the myocardium in the early and late postinfarction periods remains important. Rat left ventricular cardiomyocyte (CMC) ploidy, hypertrophy, hyperplasia, and ultrastructure were investigated in 2, 6, and 26 weeks after experimental myocardial infarction, caused by permanent ligation of left coronary artery. Cytofluorimetric study of CMC ploidy revealed no difference between normal, sham-operated, and infarcted animals for all the tested stages. However, interference microscopy indicated significant changes in cells size. CMC dry mass of infarcted rats in 2 weeks after surgery was 1.5 times lower than in control and sham operated groups. Electron microscopy analysis of CMC revealed disruption of sarcomere structure. However, in 6 weeks after surgery CMC dry mass was 1.6 times higher than in control. In 26 weeks after myocardial infarction CMC dry mass exceeded control only in peri-infarction zone. Cell counting showed that the number of left ventricular CMC, reduced as a result of myocardial infarction, was not restored during myocardial remodeling. © 2019 International Society for Advancement of Cytometry., (© 2019 International Society for Advancement of Cytometry.)
- Published
- 2019
- Full Text
- View/download PDF
626. Concise Review: Reduction of Adverse Cardiac Scarring Facilitates Pluripotent Stem Cell-Based Therapy for Myocardial Infarction.
- Author
-
Liang J, Huang W, Jiang L, Paul C, Li X, and Wang Y
- Subjects
- Animals, Cicatrix genetics, Cicatrix immunology, Cicatrix pathology, Disease Models, Animal, Fibrosis, Graft Survival drug effects, Humans, Hydrogels chemistry, Hydrogels pharmacology, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Mechanotransduction, Cellular genetics, Mechanotransduction, Cellular immunology, MicroRNAs genetics, MicroRNAs metabolism, Myocardial Infarction genetics, Myocardial Infarction immunology, Myocardial Infarction pathology, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Myofibroblasts metabolism, Myofibroblasts pathology, Paracrine Communication genetics, Paracrine Communication immunology, Pluripotent Stem Cells metabolism, Regeneration genetics, Regeneration immunology, Cicatrix prevention & control, Genetic Therapy methods, Myocardial Infarction therapy, Pluripotent Stem Cells cytology, Stem Cell Transplantation methods, Tissue Engineering methods
- Abstract
Pluripotent stem cells (PSCs) are an attractive, reliable source for generating functional cardiomyocytes for regeneration of infarcted heart. However, inefficient cell engraftment into host tissue remains a notable challenge to therapeutic success due to mechanical damage or relatively inhospitable microenvironment. Evidence has shown that excessively formed scar tissues around cell delivery sites present as mechanical and biological barriers that inhibit migration and engraftment of implanted cells. In this review, we focus on the functional responses of stem cells and cardiomyocytes during the process of cardiac fibrosis and scar formation. Survival, migration, contraction, and coupling function of implanted cells may be affected by matrix remodeling, inflammatory factors, altered tissue stiffness, and presence of electroactive myofibroblasts in the fibrotic microenvironment. Although paracrine factors from implanted cells can improve cardiac fibrosis, the transient effect is insufficient for complete repair of an infarcted heart. Furthermore, investigation of interactions between implanted cells and fibroblasts including myofibroblasts helps the identification of new targets to optimize the host substrate environment for facilitating cell engraftment and functional integration. Several antifibrotic approaches, including the use of pharmacological agents, gene therapies, microRNAs, and modified biomaterials, can prevent progression of heart failure and have been developed as adjunct therapies for stem cell-based regeneration. Investigation and optimization of new biomaterials is also required to enhance cell engraftment of engineered cardiac tissue and move PSCs from a laboratory setting into translational medicine., (© 2019 The Authors. Stem Cells published by Wiley Periodicals, Inc. on behalf of AlphaMed Press 2019.)
- Published
- 2019
- Full Text
- View/download PDF
627. Regulatory T-cells regulate neonatal heart regeneration by potentiating cardiomyocyte proliferation in a paracrine manner.
- Author
-
Li J, Yang KY, Tam RCY, Chan VW, Lan HY, Hori S, Zhou B, and Lui KO
- Subjects
- Adoptive Transfer, Aging physiology, Animals, Animals, Newborn, Cell Proliferation, Fibrosis, Forkhead Transcription Factors metabolism, Gene Expression Regulation, Developmental, Humans, Immunity, Innate, Loss of Function Mutation genetics, Macrophages metabolism, Mice, Inbred NOD, Mice, SCID, Myocardial Infarction pathology, Myocardial Infarction physiopathology, Myocytes, Cardiac metabolism, Transcriptome genetics, Up-Regulation genetics, Heart physiology, Myocytes, Cardiac cytology, Paracrine Communication, Regeneration immunology, T-Lymphocytes, Regulatory immunology
- Abstract
The neonatal mouse heart is capable of transiently regenerating after injury from postnatal day (P) 0-7 and macrophages are found important in this process. However, whether macrophages alone are sufficient to orchestrate this regeneration; what regulates cardiomyocyte proliferation; why cardiomyocytes do not proliferate after P7; and whether adaptive immune cells such as regulatory T-cells (Treg) influence neonatal heart regeneration have less studied. Methods : We employed both loss- and gain-of-function transgenic mouse models to study the role of Treg in neonatal heart regeneration. In loss-of-function studies, we treated mice with the lytic anti-CD25 antibody that specifically depletes Treg; or we treated FOXP3
DTR with diphtheria toxin that specifically ablates Treg. In gain-of-function studies, we adoptively transferred hCD2+ Treg from NOD. Foxp3hCD2 to NOD/SCID that contain Treg as the only T-cell population. Furthermore, we performed single-cell RNA-sequencing of Treg to uncover paracrine factors essential for cardiomyocyte proliferation. Results : Our results demonstrate a regenerative role of Treg in neonatal heart regeneration. Treg can directly facilitate cardiomyocyte proliferation in a paracrine manner.Conclusion : Our results demonstrate a regenerative role of Treg in neonatal heart regeneration. Treg can directly facilitate cardiomyocyte proliferation in a paracrine manner., Competing Interests: Competing Interests: The authors have declared that no competing interest exists.- Published
- 2019
- Full Text
- View/download PDF
628. Chemical suppression of specific C-C chemokine signaling pathways enhances cardiac reprogramming.
- Author
-
Guo Y, Lei I, Tian S, Gao W, Hacer K, Li Y, Wang S, Liu L, and Wang Z
- Subjects
- Actinin metabolism, Animals, Calcium metabolism, Cells, Cultured, Fibroblasts cytology, Fibroblasts metabolism, GATA4 Transcription Factor metabolism, MEF2 Transcription Factors metabolism, Mice, Myocytes, Cardiac cytology, Myocytes, Cardiac metabolism, Receptors, CCR1 metabolism, T-Box Domain Proteins metabolism, Troponin T metabolism, Cellular Reprogramming drug effects, Chemokine CCL3 metabolism, Heterocyclic Compounds, 2-Ring pharmacology, Insulin-Like Growth Factor I pharmacology, Pyrazoles pharmacology, Signal Transduction drug effects, Thiazoles pharmacology, Thiosemicarbazones pharmacology
- Abstract
Reprogramming of fibroblasts into induced cardiomyocytes (iCMs) is a potentially promising strategy for regenerating a damaged heart. However, low fibroblast-cardiomyocyte conversion rates remain a major challenge in this reprogramming. To this end, here we conducted a chemical screen and identified four agents, insulin-like growth factor-1, Mll1 inhibitor MM589, transforming growth factor-β inhibitor A83-01, and Bmi1 inhibitor PTC-209, termed IMAP, which coordinately enhanced reprogramming efficiency. Using α-muscle heavy chain-GFP-tagged mouse embryo fibroblasts as a starting cell type, we observed that the IMAP treatment increases iCM formation 6-fold. IMAP stimulated higher cardiac troponin T and α-actinin expression and increased sarcomere formation, coinciding with up-regulated expression of many cardiac genes and down-regulated fibroblast gene expression. Furthermore, IMAP promoted higher spontaneous beating and calcium transient activities of iCMs derived from neonatal cardiac fibroblasts. Intriguingly, we also observed that the IMAP treatment repressed many genes involved in immune responses, particularly those in specific C-C chemokine signaling pathways. We therefore investigated the roles of C-C motif chemokine ligand 3 (CCL3), CCL6, and CCL17 in cardiac reprogramming and observed that they inhibited iCM formation, whereas inhibitors of C-C motif chemokine receptor 1 (CCR1), CCR4, and CCR5 had the opposite effect. These results indicated that the IMAP treatment directly suppresses specific C-C chemokine signaling pathways and thereby enhances cardiac reprogramming. In conclusion, a combination of four chemicals, named here IMAP, suppresses specific C-C chemokine signaling pathways and facilitates Mef2c/Gata4/Tbx5 (MGT)-induced cardiac reprogramming, providing a potential means for iCM formation in clinical applications., (© 2019 Guo et al.)
- Published
- 2019
- Full Text
- View/download PDF
629. Advances in heart regeneration based on cardiomyocyte proliferation and regenerative potential of binucleated cardiomyocytes and polyploidization.
- Author
-
Leone M and Engel FB
- Subjects
- Animals, Cell Nucleus physiology, Humans, Polyploidy, Regenerative Medicine methods, Cell Proliferation physiology, Heart physiology, Myocytes, Cardiac physiology, Regeneration physiology
- Abstract
One great achievement in medical practice is the reduction in acute mortality of myocardial infarction due to identifying risk factors, antiplatelet therapy, optimized hospitalization and acute percutaneous coronary intervention. Yet, the prevalence of heart failure is increasing presenting a major socio-economic burden. Thus, there is a great need for novel therapies that can reverse damage inflicted to the heart. In recent years, data have accumulated suggesting that induction of cardiomyocyte proliferation might be a future option for cardiac regeneration. Here, we review the relevant literature since September 2015 concluding that it remains a challenge to verify that a therapy induces indeed cardiomyocyte proliferation. Most importantly, it is unclear that the detected increase in cardiomyocyte cell cycle activity is required for an associated improved function. In addition, we review the literature regarding the evidence that binucleated and polyploid mononucleated cardiomyocytes can divide, and put this in context to other cell types. Our analysis shows that there is significant evidence that binucleated cardiomyocytes can divide. Yet, it remains elusive whether also polyploid mononucleated cardiomyocytes can divide, how efficient proliferation of binucleated cardiomyocytes can be induced, what mechanism regulates cell cycle progression in these cells, and what fate and physiological properties the daughter cells have. In summary, we propose to standardize and independently validate cardiac regeneration studies, encourage the field to study the proliferative potential of binucleated and polyploid mononucleated cardiomyocytes, and to determine whether induction of polyploidization can enhance cardiac function post-injury., (© 2019 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2019
- Full Text
- View/download PDF
630. Excessive inflammation impairs heart regeneration in zebrafish breakdance mutant after cryoinjury.
- Author
-
Xu S, Liu C, Xie F, Tian L, Manno SH, Manno FAM 3rd, Fallah S, Pelster B, Tse G, and Cheng SH
- Subjects
- Animals, Cold Temperature adverse effects, Disease Models, Animal, Heart, Heart Injuries etiology, Inflammation etiology, Down-Regulation, Heart Injuries physiopathology, Inflammation physiopathology, Regeneration, Zebrafish physiology
- Abstract
Inflammation plays a crucial role in cardiac regeneration. Numerous advantages, including a robust regenerative ability, make the zebrafish a popular model to study cardiovascular diseases. The zebrafish breakdance (bre) mutant shares several key features with human long QT syndrome that predisposes to ventricular arrhythmias and sudden death. However, how inflammatory response and tissue regeneration following cardiac damage occur in bre mutant is unknown. Here, we have found that inflammatory response related genes were markedly expressed in the injured heart and excessive leukocyte accumulation occurred in the injured area of the bre mutant zebrafish. Furthermore, bre mutant zebrafish exhibited aberrant apoptosis and impaired heart regenerative ability after ventricular cryoinjury. Mild dosages of anti-inflammatory or prokinetic drugs protected regenerative cells from undergoing aberrant apoptosis and promoted heart regeneration in bre mutant zebrafish. We propose that immune or prokinetic therapy could be a potential therapeutic regimen for patients with genetic long QT syndrome who suffers from myocardial infarction., (Copyright © 2019. Published by Elsevier Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
631. Efficient in vivo direct conversion of fibroblasts into cardiomyocytes using a nanoparticle-based gene carrier.
- Author
-
Chang Y, Lee E, Kim J, Kwon YW, Kwon Y, and Kim J
- Subjects
- Animals, Cell Line, Cellular Reprogramming, Gold chemistry, Metal Nanoparticles chemistry, Mice, Mice, Inbred C57BL, Cellular Reprogramming Techniques methods, Fibroblasts cytology, Gene Transfer Techniques, Myocytes, Cardiac cytology
- Abstract
The reprogramming of induced cardiomyocytes (iCMs) has shown potential in regenerative medicine. However, in vivo reprogramming of iCMs is significantly inefficient, and novel gene delivery systems are required to more efficiently and safely induce in vivo reprogramming of iCMs for therapeutic applications in heart injury. In this study, we show that cationic gold nanoparticles (AuNPs) loaded with Gata4, Mef2c, and Tbx5 function as nanocarriers for cardiac reprogramming. The AuNP/GMT/PEI nanocomplexes show high reprogramming efficiency in human and mouse somatic cells with low cytotoxicity and direct conversion into iCMs without integrating factors into the genome. Importantly, AuNP/GMT/PEI nanocomplexes led to efficient in vivo conversion into cardiomyocytes after myocardial infarction (MI), resulting in the effective recovery of cardiac function and scar area. Taken together, these results show that the AuNP/GMT/PEI nanocarrier can be used to develop effective therapeutics for heart regeneration in cardiac disease patients., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
632. A long noncoding RNA NR_045363 controls cardiomyocyte proliferation and cardiac repair.
- Author
-
Wang J, Chen X, Shen D, Ge D, Chen J, Pei J, Li Y, Yue Z, Feng J, Chu M, and Nie Y
- Subjects
- Animals, Animals, Newborn, Base Sequence, Cell Proliferation, Conserved Sequence, Heart embryology, Humans, Janus Kinases metabolism, Mice, MicroRNAs metabolism, Myocardial Infarction genetics, Myocardial Infarction physiopathology, Regeneration, STAT3 Transcription Factor metabolism, Signal Transduction, Myocardium pathology, Myocytes, Cardiac cytology, Myocytes, Cardiac metabolism, RNA, Long Noncoding metabolism, Wound Healing
- Abstract
Long noncoding RNAs (lncRNAs) play important roles in the regulation of genes involved in cell proliferation. We have previously sought to more globally understand the differences of lncRNA expression between human fetal heart and adult heart to identify some functional lncRNAs which involve in the process of heart repair. We found that a highly conserved long noncoding RNA NR_045363 was mainly expressed in cardiomyocytes and rarely in non-cardiomyocytes. NR_045363 overexpression in 7-day-old mice heart could improve cardiac function and stimulate cardiomyocyte proliferation after myocardial infarction. Furthermore, NR_045363 knockdown inhibited proliferation of primary embryonic cardiomyocytes, while NR_045363 overexpression enhanced DNA synthesis and cytokinesis in neonatal cardiomyocytes in vitro. Mechanistic analysis revealed that NR_045363 promoted cardiomyocyte proliferation through interaction with miR-216a, which regulated the JAK2-STAT3 pathway. Our results showed that NR_045363 is a potent lncRNA modulator essential for cardiomyocyte proliferation., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
633. Emerging Roles for Immune Cells and MicroRNAs in Modulating the Response to Cardiac Injury.
- Author
-
Rodriguez AM and Yin VP
- Abstract
Stimulating cardiomyocyte regeneration after an acute injury remains the central goal in cardiovascular regenerative biology. While adult mammals respond to cardiac damage with deposition of rigid scar tissue, adult zebrafish and salamander unleash a regenerative program that culminates in new cardiomyocyte formation, resolution of scar tissue, and recovery of heart function. Recent studies have shown that immune cells are key to regulating pro-inflammatory and pro-regenerative signals that shift the injury microenvironment toward regeneration. Defining the genetic regulators that control the dynamic interplay between immune cells and injured cardiac tissue is crucial to decoding the endogenous mechanism of heart regeneration. In this review, we discuss our current understanding of the extent that macrophage and regulatory T cells influence cardiomyocyte proliferation and how microRNAs (miRNAs) regulate their activity in the injured heart.
- Published
- 2019
- Full Text
- View/download PDF
634. Molecular switch model for cardiomyocyte proliferation.
- Author
-
Hashmi S and Ahmad HR
- Abstract
This review deals with the human adult cardiomyocyte proliferation as a potential source for heart repair after injury. The mechanism to regain the proliferative capacity of adult cardiomyocytes is a challenge. However, recent studies are promising in showing that the 'locked' cell cycle of adult cardiomyocytes could be released through modulation of cell cycle checkpoints. In support of this are the signaling pathways of Notch, Hippo, Wnt, Akt and Jak/Stat that facilitate or inhibit the transition at cell cycle checkpoints. Cyclins and cyclin dependant kinases (CDKs) facilitate this transition which in turn is regulated by inhibitory action of pocket protein e.g. p21, p27 and p57. Transcription factors e.g. E2F, GATA4, TBx20 up regulate Cyclin A, A2, D, E, and CDK4 as promoters of cell cycle and Meis-1 and HIF-1 alpha down regulate cyclin D and E to inhibit the cell cycle. Paracrine factors like Neuregulin-1, IGF-1 and Oncostatin M and Extracellular Matrix proteins like Agrin have been involved in cardiomyocyte proliferation and dedifferentiation processes. A molecular switch model is proposed that transforms the post mitotic cell into an actively dividing cell. This model shows how the cell cycle is regulated through on- and off switch mechanisms through interaction of transcription factors and signaling pathways with proteins of the cell cycle checkpoints. Signals triggered by injury may activate the right combination of the various pathways that can 'switch on' the proliferation signals leading to myocardial regeneration.
- Published
- 2019
- Full Text
- View/download PDF
635. Recent Biomedical Applications on Stem Cell Therapy: A Brief Overview.
- Author
-
Agrawal M, Alexander A, Khan J, Giri TK, Siddique S, Dubey SK, Ajazuddin, Patel RJ, Gupta U, Saraf S, and Saraf S
- Subjects
- Cell Proliferation genetics, Humans, Wound Healing genetics, Adult Stem Cells transplantation, Cell Differentiation genetics, Embryonic Stem Cells transplantation, Regenerative Medicine trends
- Abstract
Stem cells are the specialized cell population with unique self-renewal ability and act as the precursor of all the body cells. Broadly, stem cells are of two types one is embryonic stem cells while the other is adult or somatic stem cells. Embryonic stem cells are the cells of zygote of the blastocyst which give rise to all kind of body cells including embryonic cells, and it can reconstruct a complete organism. While the adult stem cells have limited differentiation ability in comparison with embryonic stem cells and it proliferates into some specific kind of cells. This unique ability of the stem cell makes it a compelling biomedical and therapeutic tool. Stem cells primarily serve as regenerative medicine for particular tissue regeneration or the whole organ regeneration in any physical injury or disease condition (like diabetes, cancer, periodontal disorder, etc.), tissue grafting and plastic surgery, etc. Along with this, it is also used in various preclinical and clinical investigations, biomedical engineering and as a potential diagnostic tool (such as the development of biomarkers) for non-invasive diagnosis of severe disorders. In this review article, we have summarized the application of stem cell as regenerative medicine and in the treatment of various chronic diseases., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.net.)
- Published
- 2019
- Full Text
- View/download PDF
636. Therapeutic Potential of Pluripotent Stem Cells for Cardiac Repair after Myocardial Infarction.
- Author
-
Okano S and Shiba Y
- Subjects
- Animals, Cell Differentiation, Humans, Myocardial Infarction therapy, Myocytes, Cardiac transplantation, Pluripotent Stem Cells cytology
- Abstract
Myocardial infarction occurs as a result of acute arteriosclerotic plaque rupture in the coronary artery, triggering strong inflammatory responses. The necrotic cardiomyocytes are gradually replaced with noncontractile scar tissue that eventually manifests as heart failure. Pluripotent stem cells (PSCs) show great promise for widespread clinical applications, particularly for tissue regeneration, and are being actively studied around the world to help elucidate disease mechanisms and in the development of new drugs. Human induced PSCs also show potential for regeneration of the myocardial tissue in experiments with small animals and in in vitro studies. Although emerging evidence points to the effectiveness of these stem cell-derived cardiomyocytes in cardiac regeneration, several challenges remain before clinical application can become a reality. Here, we provide an overview of the present state of PSC-based heart regeneration and highlight the remaining hurdles, with a particular focus on graft survival, immunogenicity, posttransplant arrhythmia, maintained function, and tumor formation. Rapid progress in this field along with advances in biotechnology are expected to resolve these issues, which will require international collaboration and standardization.
- Published
- 2019
- Full Text
- View/download PDF
637. The Structure of the Periostin Gene, Its Transcriptional Control and Alternative Splicing, and Protein Expression.
- Author
-
Kudo A
- Subjects
- Humans, Alternative Splicing, Cell Adhesion Molecules genetics
- Abstract
Although many studies have described the role of periostin in various diseases, the functions of periostin derived from alternative splicing and proteinase cleavage at its C-terminus remain unknown. Further experiments investigating the periostin structures that are relevant to diseases are essential for an in-depth understanding of their functions, which would accelerate their clinical applications by establishing new approaches for curing intractable diseases. Furthermore, this understanding would enhance our knowledge of novel functions of periostin related to stemness and response to mechanical stress .
- Published
- 2019
- Full Text
- View/download PDF
638. Regenerative Medicine and Biomarkers for Dilated Cardiomyopathy
- Author
-
Lesizza P, Aleksova A, Ortis B, Beltrami AP, Giacca M, Sinagra G, Sinagra G, Merlo M, and Pinamonti B
- Abstract
Dilated cardiomyopathy is characterized by progressive cardiomyocyte loss leading to ventricle dilation and dysfunction. Over the last decade, multiple evidence has shown that treatment of this condition might be attempted through the administration of either cells of various derivations or nucleic acids. In the case of cell therapy, there is ample consensus that no stem cells can directly regenerate the myocardium; however some cell types could provide benefit through a paracrine function on resident cardiomyocytes. Various nucleic acids, including microRNAs and antisense locked nucleic acids targeting microRNAs and long non-coding RNAs, can stimulate regeneration by promoting the proliferation potential of endogenous cardiomyocytes. Albeit at the preclinical phase, these approaches hold a great promise for the development of innovative therapeutics. Patients with idiopathic dilated cardiomyopathy are generally young subjects. Therefore, the assessment of prognosis is essential. Biomarkers are nowadays widely available and are useful tools for risk stratification. Besides HF-dedicated biomarkers, such as natriuretic peptides, galectin-3, soluble ST2 and troponins, also the evaluation of inflammatory response (interleukins, growth factors), renal function (NGAL, KIM-1) and anaemia are particularly important for a correct prognostic stratification. Moreover, when all of these biomarkers are used and combined in a multimarker model, the prediction of prognosis becomes more accurate, reflecting the importance of a holistic evaluation of patients., (Copyright 2019, The Author(s).)
- Published
- 2019
- Full Text
- View/download PDF
639. The regulation and function of the Hippo pathway in heart regeneration.
- Author
-
Liu S and Martin JF
- Subjects
- Cell Proliferation genetics, Cell Self Renewal genetics, Heart growth & development, Heart physiopathology, Heart Failure genetics, Heart Failure physiopathology, Hippo Signaling Pathway, Humans, Myocytes, Cardiac pathology, Protein Serine-Threonine Kinases antagonists & inhibitors, Regeneration physiology, Signal Transduction genetics, Adult Stem Cells transplantation, Heart Failure therapy, Protein Serine-Threonine Kinases genetics, Regeneration genetics
- Abstract
Heart failure caused by cardiomyocyte loss and fibrosis is a leading cause of death worldwide. Although current treatments for heart failure such as heart transplantation and left ventricular assist device implantation have obvious value, new approaches are needed. Endogenous adult cardiomyocyte renewal is measurable but inefficient and inadequate in response to extensive acute heart damage. Stimulating self-renewal of endogenous cardiomyocytes holds great promise for heart repair. Uncovering the genetic mechanisms underlying cardiomyocyte renewal is a critical step in developing new approaches to repairing the heart. Recent studies have revealed that the inhibition of the Hippo pathway is sufficient to promote the proliferation of endogenous cardiomyocytes, indicating that the manipulation of the Hippo pathway in the heart may be a promising treatment for heart failure in the future. We summarize recent findings that have shed light on the function of the Hippo pathway in heart regeneration. We also discuss the mechanisms by which Hippo pathway inhibition promotes heart regeneration and how the Hippo pathway responds to different types of injury or stress during the regenerative process. This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration., (© 2018 Wiley Periodicals, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
640. Decoding the Heart through Next Generation Sequencing Approaches.
- Author
-
Pawlak, Michal, Niescierowicz, Katarzyna, and Winata, Cecilia Lanny
- Subjects
- *
CONGENITAL heart disease , *HEART development , *RNA sequencing , *HEART cells , *TRANSCRIPTION factors - Abstract
Vertebrate organs develop through a complex process which involves interaction between multiple signaling pathways at the molecular, cell, and tissue levels. Heart development is an example of such complex process which, when disrupted, results in congenital heart disease (CHD). This complexity necessitates a holistic approach which allows the visualization of genome-wide interaction networks, as opposed to assessment of limited subsets of factors. Genomics offers a powerful solution to address the problem of biological complexity by enabling the observation of molecular processes at a genome-wide scale. The emergence of next generation sequencing (NGS) technology has facilitated the expansion of genomics, increasing its output capacity and applicability in various biological disciplines. The application of NGS in various aspects of heart biology has resulted in new discoveries, generating novel insights into this field of study. Here we review the contributions of NGS technology into the understanding of heart development and its disruption reflected in CHD and discuss how emerging NGS based methodologies can contribute to the further understanding of heart repair. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
641. Delineating the Dynamic Transcriptome Response of mRNA and microRNA during Zebrafish Heart Regeneration.
- Author
-
Klett H, Jürgensen L, Most P, Busch M, Günther F, Dobreva G, Leuschner F, Hassel D, Busch H, and Boerries M
- Subjects
- Animals, Cell Differentiation, Cell Line, Cell Proliferation, Cluster Analysis, Myocytes, Cardiac cytology, Myocytes, Cardiac metabolism, Principal Component Analysis, Rats, Regeneration, Zebrafish, Heart physiology, MicroRNAs metabolism, RNA, Messenger metabolism, Transcriptome
- Abstract
Heart diseases are the leading cause of death for the vast majority of people around the world, which is often due to the limited capability of human cardiac regeneration. In contrast, zebrafish have the capacity to fully regenerate their hearts after cardiac injury. Understanding and activating these mechanisms would improve health in patients suffering from long-term consequences of ischemia. Therefore, we monitored the dynamic transcriptome response of both mRNA and microRNA in zebrafish at 1⁻160 days post cryoinjury (dpi). Using a control model of sham-operated and healthy fish, we extracted the regeneration specific response and further delineated the spatio-temporal organization of regeneration processes such as cell cycle and heart function. In addition, we identified novel (miR-148/152, miR-218b and miR-19) and previously known microRNAs among the top regulators of heart regeneration by using theoretically predicted target sites and correlation of expression profiles from both mRNA and microRNA. In a cross-species effort, we validated our findings in the dynamic process of rat myoblasts differentiating into cardiomyocytes-like cells (H9c2 cell line). Concluding, we elucidated different phases of transcriptomic responses during zebrafish heart regeneration. Furthermore, microRNAs showed to be important regulators in cardiomyocyte proliferation over time.
- Published
- 2018
- Full Text
- View/download PDF
642. Covering and Re-Covering the Heart: Development and Regeneration of the Epicardium.
- Author
-
Cao Y and Cao J
- Abstract
The epicardium, a mesothelial layer that envelops vertebrate hearts, has become a therapeutic target in cardiac repair strategies because of its vital role in heart development and cardiac injury response. Epicardial cells serve as a progenitor cell source and signaling center during both heart development and regeneration. The importance of the epicardium in cardiac repair strategies has been reemphasized by recent progress regarding its requirement for heart regeneration in zebrafish, and by the ability of patches with epicardial factors to restore cardiac function following myocardial infarction in mammals. The live surveillance of epicardial development and regeneration using zebrafish has provided new insights into this topic. In this review, we provide updated knowledge about epicardial development and regeneration.
- Published
- 2018
- Full Text
- View/download PDF
643. Genetic and epigenetic regulation of cardiomyocytes in development, regeneration and disease.
- Author
-
Cui M, Wang Z, Bassel-Duby R, and Olson EN
- Subjects
- Animals, Gene Expression Regulation, Developmental, Humans, Epigenesis, Genetic, Heart embryology, Heart Diseases genetics, Myocytes, Cardiac metabolism, Regeneration genetics
- Abstract
Embryonic and postnatal life depend on the uninterrupted function of cardiac muscle cells. These cells, termed cardiomyocytes, display many fascinating behaviors, including complex morphogenic movements, interactions with other cell types of the heart, persistent contractility and quiescence after birth. Each of these behaviors depends on complex interactions between both cardiac-restricted and widely expressed transcription factors, as well as on epigenetic modifications. Here, we review recent advances in our understanding of the genetic and epigenetic control of cardiomyocyte differentiation and proliferation during heart development, regeneration and disease. We focus on those regulators that are required for both heart development and disease, and highlight the regenerative principles that might be manipulated to restore function to the injured adult heart., Competing Interests: Competing interestsE.N.O. is a co-founder and member of the Scientific Advisory Board of Tenaya Therapeutics and holds equity in the company., (© 2018. Published by The Company of Biologists Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
644. Coronary Vasculature in Cardiac Development and Regeneration.
- Author
-
Kapuria S, Yoshida T, and Lien CL
- Abstract
Functional coronary circulation is essential for a healthy heart in warm-blooded vertebrates, and coronary diseases can have a fatal consequence. Despite the growing interest, the knowledge about the coronary vessel development and the roles of new coronary vessel formation during heart regeneration is still limited. It is demonstrated that early revascularization is required for efficient heart regeneration. In this comprehensive review, we first describe the coronary vessel formation from an evolutionary perspective. We further discuss the cell origins of coronary endothelial cells and perivascular cells and summarize the critical signaling pathways regulating coronary vessel development. Lastly, we focus on the current knowledge about the molecular mechanisms regulating heart regeneration in zebrafish, a genetically tractable vertebrate model with a regenerative adult heart and well-developed coronary system., Competing Interests: The authors declare no conflict of interest.
- Published
- 2018
- Full Text
- View/download PDF
645. Design and formulation of functional pluripotent stem cell-derived cardiac microtissues
- Author
-
Stéphane Massé, Milica Radisic, Craig A. Simmons, Gordon Keller, Vikram Deshpande, Nimalan Thavandiran, Peter W. Zandstra, Bogdan M. Beca, Alexander Mikryukov, Kumaraswamy Nanthakumar, Nicole Dubois, J. Patrick McGarry, and Christopher S. Chen
- Subjects
cardiac toxicity ,Pluripotent Stem Cells ,heart regeneration ,actin cytoskeleton ,Finite Element Analysis ,cardiomyocytes ,heart ,Biology ,contractility ,Corrections ,Homeobox protein Nkx-2.5 ,resistance ,Tissue engineering ,Humans ,CD90 ,arrhythmia disease model ,Induced pluripotent stem cell ,microfabrication ,Homeodomain Proteins ,model ,Multidisciplinary ,Tissue Engineering ,Cluster of differentiation ,Cardiac electrophysiology ,Myocardium ,In vitro toxicology ,Human heart ,tissue ,differentiation ,simulation ,Electric Stimulation ,Biomechanical Phenomena ,Cellular Microenvironment ,PNAS Plus ,Homeobox Protein Nkx-2.5 ,Thy-1 Antigens ,stress-fiber ,Transcription Factors ,Biomedical engineering - Abstract
Access to robust and information-rich human cardiac tissue models would accelerate drug-based strategies for treating heart disease. Despite significant effort, the generation of high-fidelity adult-like human cardiac tissue analogs remains challenging. We used computational modeling of tissue contraction and assembly mechanics in conjunction with microfabricated constraints to guide the design of aligned and functional 3D human pluripotent stem cell (hPSC)-derived cardiac microtissues that we term cardiac microwires (CMWs). Miniaturization of the platform circumvented the need for tissue vascularization and enabled higher-throughput image-based analysis of CMW drug responsiveness. CMW tissue properties could be tuned using electromechanical stimuli and cell composition. Specifically, controlling self-assembly of 3D tissues in aligned collagen, and pacing with point stimulation electrodes, were found to promote cardiac maturation-associated gene expression and in vivo-like electrical signal propagation. Furthermore, screening a range of hPSC-derived cardiac cell ratios identified that 75% NKX2 Homeobox 5 (NKX2-5)+ cardiomyocytes and 25% Cluster of Differentiation 90 OR (CD90)+ nonmyocytes optimized tissue remodeling dynamics and yielded enhanced structural and functional properties. Finally, we demonstrate the utility of the optimized platform in a tachycardic model of arrhythmogenesis, an aspect of cardiac electrophysiology not previously recapitulated in 3D in vitro hPSC-derived cardiac microtissue models. The design criteria identified with our CMW platform should accelerate the development of predictive in vitro assays of human heart tissue function.
- Published
- 2013
646. The Origin of Human Mesenchymal Stromal Cells Dictates Their Reparative Properties
- Author
-
Natalie Landa-Rouben, David Kain, Eyal Winkler, Dov Zipori, Ayelet Itzhaki-Alfia, Ehud Raanani, Meirav Pevsner-Fischer, Nili Naftali-Shani, Radka Holbova, Jonathan Leor, Ariel Tessone, Eran Millet, Jens Kastrup, Natali Molotski, Avishay Grupper, Shimrit Adutler-Lieber, Micha S. Feinberg, and Elad Asher
- Subjects
endocrine system ,medicine.medical_specialty ,heart regeneration ,epicardial fat ,Myocardial Infarction ,Myocardial Ischemia ,Adipose tissue ,030204 cardiovascular system & hematology ,Mesenchymal Stem Cell Transplantation ,Proinflammatory cytokine ,mesenchymal stromal/stem cells ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Animals ,Humans ,Medicine ,Pericardium ,cardiovascular diseases ,Myocardial infarction ,Atrium (heart) ,Ventricular remodeling ,Cells, Cultured ,Original Research ,030304 developmental biology ,Heart Failure ,0303 health sciences ,business.industry ,Myocardium ,Mesenchymal stem cell ,Heart ,Mesenchymal Stem Cells ,equipment and supplies ,medicine.disease ,adipose tissue ,macrophages ,Transplantation ,medicine.anatomical_structure ,inflammation ,cardiovascular system ,Cardiology ,Cardiology and Cardiovascular Medicine ,business - Abstract
Background Human mesenchymal stromal cells ( hMSC s) from adipose cardiac tissue have attracted considerable interest in regard to cell‐based therapies. We aimed to test the hypothesis that hMSC s from the heart and epicardial fat would be better cells for infarct repair. Methods and Results We isolated and grew hMSC s from patients with ischemic heart disease from 4 locations: epicardial fat, pericardial fat, subcutaneous fat, and the right atrium. Significantly, hMSC s from the right atrium and epicardial fat secreted the highest amounts of trophic and inflammatory cytokines, while hMSC s from pericardial and subcutaneous fat secreted the lowest. Relative expression of inflammation‐ and fibrosis‐related genes was considerably higher in hMSC s from the right atrium and epicardial fat than in subcutaneous fat hMSC s. To determine the functional effects of hMSC s, we allocated rats to hMSC transplantation 7 days after myocardial infarction. Atrial hMSC s induced greatest infarct vascularization as well as highest inflammation score 27 days after transplantation. Surprisingly, cardiac dysfunction was worst after transplantation of hMSC s from atrium and epicardial fat and minimal after transplantation of hMSC s from subcutaneous fat. These findings were confirmed by using hMSC transplantation in immunocompromised mice after myocardial infarction. Notably, there was a correlation between tumor necrosis factor‐α secretion from hMSC s and posttransplantation left ventricular remodeling and dysfunction. Conclusions Because of their proinflammatory properties, hMSC s from the right atrium and epicardial fat of cardiac patients could impair heart function after myocardial infarction. Our findings might be relevant to autologous mesenchymal stromal cell therapy and development and progression of ischemic heart disease.
- Published
- 2013
647. Special Issue 11: Cardiac Stem Cells
- Author
-
Cyganek, Lukas, Chen, Simin, Borchert, Thomas, and Guan, Kaomei
- Subjects
Cardiac progenitor cells ,Cardiomyocytes ,Molecular mechanism ,Cardiac resident stem cells ,Heart regeneration - Abstract
Heart disease is the principal cause of death in humans. Stem cell-based therapy for heart regeneration has long been seen as a potential application since the heart lacks adequate intrinsic regenerative potential. In the cardiovascular field, clinical trials have already been carried out by implantation of both bone marrow-derived stem cells and cardiac resident progenitor cells derived from the adult heart tissue into the injured myocardium to restore the functionality of the heart after damage. However, before a robust stem and progenitor cell-based therapy for cardiovascular diseases can be applied in the clinical setting, more research is necessary to generate sufficient quantities of functional cardiomyocytes from stem cells and to understand behavior of cardiomyocytes upon transplantation. A comprehensive understanding of the developmental processes involved in cardiogenesis might support further investigations in more efficient cell-based regeneration therapies. This review discusses the molecular aspects of cardiogenesis during early development and links the insights with the in vitro generation of cardiac progenitor cells as well as functional cardiomyocytes. Furthermore, we discuss the advantages of cardiac progenitor cells and cardiomyocytes derived from pluripotent stem cells, cardiac resident stem cells in regenerative applications to cope with the damaged heart. Open-Access-Publikationsfonds 2013 peerReviewed
- Published
- 2013
648. Collagen XII Contributes to Epicardial and Connective Tissues in the Zebrafish Heart during Ontogenesis and Regeneration
- Author
-
Thomas Bise, Jan Marro, Catherine Pfefferli, Anne-Sophie de Preux Charles, and Anna Jaźwińska
- Subjects
0301 basic medicine ,Cell signaling ,Pathology ,lcsh:Medicine ,Signal transduction ,Biochemistry ,Animals, Genetically Modified ,Extracellular matrix ,0302 clinical medicine ,Transforming Growth Factor beta ,Heart Regeneration ,Morphogenesis ,Medicine and Health Sciences ,lcsh:Science ,Zebrafish ,In Situ Hybridization ,Multidisciplinary ,biology ,Fishes ,Signaling cascades ,Heart ,Animal Models ,Epicardium ,Up-Regulation ,Extracellular Matrix ,Cell biology ,medicine.anatomical_structure ,Connective Tissue ,Osteichthyes ,Benzamides ,Vertebrates ,Anatomy ,Cellular Structures and Organelles ,Pericardium ,Type I collagen ,Research Article ,Collagen Type XII ,medicine.medical_specialty ,Heart Ventricles ,Connective tissue ,Dioxoles ,Research and Analysis Methods ,Collagen Type I ,03 medical and health sciences ,Model Organisms ,medicine ,Animals ,Regeneration ,Vimentin ,cardiovascular diseases ,Fibroblast ,Myocardium ,Regeneration (biology) ,lcsh:R ,Organisms ,Biology and Life Sciences ,Proteins ,Cell Biology ,Zebrafish Proteins ,biology.organism_classification ,Collagen, type I, alpha 1 ,030104 developmental biology ,Microscopy, Fluorescence ,TGF-beta signaling cascade ,Connective tissue metabolism ,Cardiovascular Anatomy ,lcsh:Q ,Receptors, Transforming Growth Factor beta ,Collagens ,Organism Development ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Zebrafish heart regeneration depends on cardiac cell proliferation, epicardium activation and transient reparative tissue deposition. The contribution and the regulation of specific collagen types during the regenerative process, however, remain poorly characterized. Here, we identified that the non-fibrillar type XII collagen, which serves as a matrix-bridging component, is expressed in the epicardium of the zebrafish heart, and is boosted after cryoinjury-induced ventricular damage. During heart regeneration, an intense deposition of Collagen XII covers the outer epicardial cap and the interstitial reparative tissue. Analysis of the activated epicardium and fibroblast markers revealed a heterogeneous cellular origin of Collagen XII. Interestingly, this matrix-bridging collagen co-localized with fibrillar type I collagen and several glycoproteins in the post-injury zone, suggesting its role in tissue cohesion. Using SB431542, a selective inhibitor of the TGF-β receptor, we showed that while the inhibitor treatment did not affect the expression of collagen 12 and collagen 1a2 in the epicardium, it completely suppressed the induction of both genes in the fibrotic tissue. This suggests that distinct mechanisms might regulate collagen expression in the outer heart layer and the inner injury zone. On the basis of this study, we postulate that the TGF-β signaling pathway induces and coordinates formation of a transient collagenous network that comprises fibril-forming Collagen I and fiber-associated Collagen XII, both of which contribute to the reparative matrix of the regenerating zebrafish heart.
- Published
- 2016
649. Neuregulin-1 Administration Protocols Sufficient for Stimulating Cardiac Regeneration in Young Mice Do Not Induce Somatic, Organ, or Neoplastic Growth
- Author
-
Balakrishnan Ganapathy, Nikitha Nandhagopal, Yijen L. Wu, David A. Bennett, Bernhard Kühn, Brian D. Polizzotti, and Alparslan Asan
- Subjects
0301 basic medicine ,Aging ,Pathology ,Physiology ,Receptor expression ,medicine.medical_treatment ,Respiratory System ,lcsh:Medicine ,030204 cardiovascular system & hematology ,Kidney ,Diagnostic Radiology ,Mice ,Subcutaneous injection ,Mathematical and Statistical Techniques ,0302 clinical medicine ,Neoplasms ,Heart Regeneration ,Morphogenesis ,Medicine and Health Sciences ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,lcsh:Science ,Routes of Administration ,education.field_of_study ,Multidisciplinary ,biology ,Radiology and Imaging ,Heart ,Organ Size ,Magnetic Resonance Imaging ,Recombinant Proteins ,3. Good health ,ErbB Receptors ,medicine.anatomical_structure ,Physiological Parameters ,Physical Sciences ,Anatomy ,Statistics (Mathematics) ,Research Article ,medicine.medical_specialty ,Imaging Techniques ,Neuregulin-1 ,Population ,Spleen ,Research and Analysis Methods ,03 medical and health sciences ,Diagnostic Medicine ,Internal medicine ,medicine ,Animals ,Humans ,Regeneration ,Statistical Methods ,Neuregulin 1 ,education ,Pharmacology ,Analysis of Variance ,business.industry ,Ribosomal Protein S6 Kinases ,Growth factor ,Body Weight ,lcsh:R ,Biology and Life Sciences ,Kidneys ,Renal System ,medicine.disease ,030104 developmental biology ,Endocrinology ,Animals, Newborn ,Gene Expression Regulation ,Subcutaneous Injections ,Heart failure ,Cardiovascular Anatomy ,biology.protein ,lcsh:Q ,Lungs ,business ,Organism Development ,Mathematics ,Developmental Biology - Abstract
Background We previously developed and validated a strategy for stimulating heart regeneration by administration of recombinant neuregulin (rNRG1), a growth factor, in mice. rNRG1 stimulated proliferation of heart muscle cells, cardiomyocytes, and was most effective when administration began during the neonatal period. Our results suggested the use of rNRG1 to treat pediatric patients with heart failure. However, administration in this age group may stimulate growth outside of the heart. Methods NRG1 and ErbB receptor expression was determined by RT-PCR. rNRG1 concentrations in serum were quantified by ELISA. Mice that received protocols of recombinant neuregulin1-β1 administration (rNRG1, 100 ng/g body weight, daily subcutaneous injection for the first month of life), previously shown to induce cardiac regeneration, were examined at pre-determined intervals. Somatic growth was quantified by weighing. Organ growth was quantified by MRI and by weighing. Neoplastic growth was examined by MRI, visual inspection, and histopathological analyses. Phospho-ERK1/2 and S6 kinase were analyzed with Western blot and ELISA, respectively. Results Lung, spleen, liver, kidney, brain, and breast gland exhibited variable expression of the NRG1 receptors ErbB2, ErbB3, ErbB4, and NRG1. Body weight and tibia length were not altered in mice receiving rNRG1. MRI showed that administration of rNRG1 did not alter the volume of the lungs, liver, kidneys, brain, or spinal cord. Administration of rNRG1 did not alter the weight of the lungs, spleen, liver, kidneys, or brain. MRI, visual inspection, and histopathological analyses showed no neoplastic growth. Follow-up for 6 months showed no alteration of somatic or organ growth. rNRG1 treatment increased the levels of phospho-ERK1/2, but not phospho-S6 kinase. Conclusions Administration protocols of rNRG1 for stimulating cardiac regeneration in mice during the first month of life did not induce unwanted growth effects. Further studies may be required to determine whether this is the case in a corresponding human population.
- Published
- 2016
650. PhOTO Zebrafish: A Transgenic Resource for In Vivo Lineage Tracing during Development and Regeneration
- Author
-
William P. Dempsey, Scott E. Fraser, and Periklis Pantazis
- Subjects
Embryo, Nonmammalian ,Cell division ,Cerulean ,Animals, Genetically Modified ,LIGHT-SHEET MICROSCOPY ,Zebrafish ,Animal Management ,Multidisciplinary ,biology ,Systems Biology ,EMBRYO ,PROLIFERATION ,Agriculture ,Animal Models ,Complex cell ,Cell biology ,Multidisciplinary Sciences ,medicine.anatomical_structure ,Animal Fins ,Medicine ,Science & Technology - Other Topics ,Stem cell ,Genetic Engineering ,Cell Division ,HEART REGENERATION ,Research Article ,Biotechnology ,General Science & Technology ,Science ,FIN REGENERATION ,Genetic Vectors ,Model Organisms ,MD Multidisciplinary ,medicine ,Animals ,Regeneration ,Cell Lineage ,Nuclear membrane ,Biology ,Cell Nucleus ,Science & Technology ,Regeneration (biology) ,DEDIFFERENTIATION ,Cell Membrane ,Gastrulation ,PLANE ILLUMINATION ,biology.organism_classification ,CELLS ,BLASTEMA ,Organism Development ,SYSTEM ,Developmental Biology - Abstract
Background Elucidating the complex cell dynamics (divisions, movement, morphological changes, etc.) underlying embryonic development and adult tissue regeneration requires an efficient means to track cells with high fidelity in space and time. To satisfy this criterion, we developed a transgenic zebrafish line, called PhOTO, that allows photoconvertible optical tracking of nuclear and membrane dynamics in vivo. Methodology PhOTO zebrafish ubiquitously express targeted blue fluorescent protein (FP) Cerulean and photoconvertible FP Dendra2 fusions, allowing for instantaneous, precise targeting and tracking of any number of cells using Dendra2 photoconversion while simultaneously monitoring global cell behavior and morphology. Expression persists through adulthood, making the PhOTO zebrafish an excellent tool for studying tissue regeneration: after tail fin amputation and photoconversion of a ∼100µm stripe along the cut area, marked differences seen in how cells contribute to the new tissue give detailed insight into the dynamic process of regeneration. Photoconverted cells that contributed to the regenerate were separated into three distinct populations corresponding to the extent of cell division 7 days after amputation, and a subset of cells that divided the least were organized into an evenly spaced, linear orientation along the length of the newly regenerating fin. Conclusions/Significance PhOTO zebrafish have wide applicability for lineage tracing at the systems-level in the early embryo as well as in the adult, making them ideal candidate tools for future research in development, traumatic injury and regeneration, cancer progression, and stem cell behavior., PLoS ONE, 7 (3), ISSN:1932-6203
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.