999 results on '"heart regeneration"'
Search Results
402. In vivo monitoring of cardiomyocyte proliferation to identify chemical modifiers of heart regeneration.
- Author
-
Wen-Yee Choi, Gemberling, Matthew, Jinhu Wang, Holdway, Jennifer E., Meng-Chieh Shen, Karlstrom, Rolf O., and Poss, Kenneth D.
- Subjects
- *
BONE morphogenetic proteins , *MYOCARDIAL infarction , *HYPOGLYCEMIC agents , *CORONARY disease , *BLOOD circulation disorders , *PANCREATIC secretions - Abstract
Adult mammalian cardiomyocytes have little capacity to proliferate in response to injury, a deficiency that underlies the poor regenerative ability of human hearts after myocardial infarction. By contrast, zebrafish regenerate heart muscle after trauma by inducing proliferation of spared cardiomyocytes, providing a model for identifying manipulations that block or enhance these events. Although direct genetic or chemical screens of heart regeneration in adult zebrafish present several challenges, zebrafish embryos are ideal for high-throughput screening. Here, to visualize cardiomyocyte proliferation events in live zebrafish embryos, we generated transgenic zebrafish lines that employ fluorescent ubiquitylation-based cell cycle indicator (FUCCI) technology. We then performed a chemical screen and identified several small molecules that increase or reduce cardiomyocyte proliferation during heart development. These compounds act via Hedgehog, Insulin-like growth factor or Transforming growth factor β-signaling pathways. Direct examination of heart regeneration after mechanical or genetic ablation injuries indicated that these pathways are activated in regenerating cardiomyocytes and that they can be pharmacologically manipulated to inhibit or enhance cardiomyocyte proliferation during adult heart regeneration. Our findings describe a new screening system that identifies molecules and pathways with the potential to modify heart regeneration. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
403. Prolonged Myocardial Regenerative Capacity in Neonatal Opossum.
- Author
-
Nishiyama C, Saito Y, Sakaguchi A, Kaneko M, Kiyonari H, Xu Y, Arima Y, Uosaki H, and Kimura W
- Subjects
- Animals, Animals, Newborn, Cell Proliferation, Mice, Myocytes, Cardiac metabolism, AMP-Activated Protein Kinases metabolism, Heart physiology, Monodelphis, Myocardial Infarction metabolism, Regeneration
- Abstract
Background: Early neonates of both large and small mammals are able to regenerate the myocardium through cardiomyocyte proliferation for only a short period after birth. This myocardial regenerative capacity declines in parallel with withdrawal of cardiomyocytes from the cell cycle in the first few postnatal days. No mammalian species examined to date has been found capable of a meaningful regenerative response to myocardial injury later than 1 week after birth., Methods: We examined cardiomyocyte proliferation in neonates of the marsupial opossum ( Monodelphis domestica ) by immunostaining at various times after birth. The regenerative capacity of the postnatal opossum myocardium was assessed after either apex resection or induction of myocardial infarction at postnatal day 14 or 29, whereas that of the postnatal mouse myocardium was assessed after myocardial infarction at postnatal day 7. Bioinformatics data analysis, immunofluorescence staining, and pharmacological and genetic intervention were applied to determine the role of AMPK (5'-AMP-activated protein kinase) signaling in regulation of the mammalian cardiomyocyte cell cycle., Results: Opossum neonates were found to manifest cardiomyocyte proliferation for at least 2 weeks after birth at a frequency similar to that apparent in early neonatal mice. Moreover, the opossum heart at postnatal day 14 showed substantial regenerative capacity both after apex resection and after myocardial infarction injury, whereas this capacity had diminished by postnatal day 29. Transcriptomic and immunofluorescence analyses indicated that AMPK signaling is activated in postnatal cardiomyocytes of both opossum and mouse. Pharmacological or genetic inhibition of AMPK signaling was sufficient to extend the postnatal window of cardiomyocyte proliferation in both mouse and opossum neonates as well as of cardiac regeneration in neonatal mice., Conclusions: The marsupial opossum maintains cardiomyocyte proliferation and a capacity for myocardial regeneration for at least 2 weeks after birth. As far as we are aware, this is the longest postnatal duration of such a capacity among mammals examined to date. AMPK signaling was implicated as an evolutionarily conserved regulator of mammalian postnatal cardiomyocyte proliferation.
- Published
- 2022
- Full Text
- View/download PDF
404. Metabolic Regulation of Cardiac Regeneration.
- Author
-
Duan X, Liu X, and Zhan Z
- Abstract
The mortality due to heart diseases remains highest in the world every year, with ischemic cardiomyopathy being the prime cause. The irreversible loss of cardiomyocytes following myocardial injury leads to compromised contractility of the remaining myocardium, adverse cardiac remodeling, and ultimately heart failure. The hearts of adult mammals can hardly regenerate after cardiac injury since adult cardiomyocytes exit the cell cycle. Nonetheless, the hearts of early neonatal mammals possess a stronger capacity for regeneration. To improve the prognosis of patients with heart failure and to find the effective therapeutic strategies for it, it is essential to promote endogenous regeneration of adult mammalian cardiomyocytes. Mitochondrial metabolism maintains normal physiological functions of the heart and compensates for heart failure. In recent decades, the focus is on the changes in myocardial energy metabolism, including glucose, fatty acid, and amino acid metabolism, in cardiac physiological and pathological states. In addition to being a source of energy, metabolites are becoming key regulators of gene expression and epigenetic patterns, which may affect heart regeneration. However, the myocardial energy metabolism during heart regeneration is majorly unknown. This review focuses on the role of energy metabolism in cardiac regeneration, intending to shed light on the strategies for manipulating heart regeneration and promoting heart repair after cardiac injury., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Duan, Liu and Zhan.)
- Published
- 2022
- Full Text
- View/download PDF
405. Cardiac Reprogramming Factors Synergistically Activate Genome-wide Cardiogenic Stage-Specific Enhancers.
- Author
-
Hashimoto, Hisayuki, Hashimoto, Hisayuki, Wang, Zhaoning, Garry, Glynnis A, Malladi, Venkat S, Botten, Giovanni A, Ye, Wenduo, Zhou, Huanyu, Osterwalder, Marco, Dickel, Diane E, Visel, Axel, Liu, Ning, Bassel-Duby, Rhonda, Olson, Eric N, Hashimoto, Hisayuki, Hashimoto, Hisayuki, Wang, Zhaoning, Garry, Glynnis A, Malladi, Venkat S, Botten, Giovanni A, Ye, Wenduo, Zhou, Huanyu, Osterwalder, Marco, Dickel, Diane E, Visel, Axel, Liu, Ning, Bassel-Duby, Rhonda, and Olson, Eric N
- 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.
- Published
- 2019
406. Migration of cardiomyocytes is essential for heart regeneration in zebrafish.
- Author
-
Itou, Junji, Oishi, Isao, Kawakami, Hiroko, Glass, Tiffany J., Richter, Jenna, Johnson, Austin, Lund, Troy C., and Kawakami, Yasuhiko
- Subjects
- *
ZEBRA danio , *HEART physiology , *HEART injuries , *HEART cells , *EMIGRATION & immigration ,HEART hemorrhage - Abstract
The article presents information regarding the role of the migration of cardiomyocytes in the zebrafish heart regeneration. It mentions that the Adult zebrafish can regenerate injured heart tissue by proliferating pre-existing cardiomyocytes. It also informs that due this capability of zebrafish, it can be served as model to study the repairing of injured heart.
- Published
- 2012
- Full Text
- View/download PDF
407. Cardiac Regenerative Capacity and Mechanisms.
- Author
-
Kikuchi, Kazu and Poss, Kenneth D.
- Subjects
- *
CARDIAC regeneration , *CORONARY circulation , *MYOCARDIAL infarction , *HEART failure , *CELL differentiation , *HEART cells , *PROGENITOR cells - Abstract
The heart holds the monumental yet monotonous task of maintaining circulation. Although cardiac function is critical to other organs and to life itself, mammals are not equipped with significant natural capacity to replace heart muscle that has been lost by injury. This deficiency plays a role in leaving millions worldwide vulnerable to heart failure each year. By contrast, certain other vertebrate species such as zebrafish are strikingly good at heart regeneration. A cellular and molecular understanding of endogenous regenerative mechanisms and advances in methodology to transplant cells together project a future in which cardiac muscle regeneration can be therapeutically stimulated in injured human hearts. This review focuses on what has been discovered recently about cardiac regenerative capacity and how natural mechanisms of heart regeneration in model systems are stimulated and maintained. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
408. Heart regeneration.
- Author
-
Kozar-Kamińska, Katarzyna
- Subjects
- *
HEART failure , *HEART disease related mortality , *ETIOLOGY of diseases , *CARDIAC regeneration , *CELLULAR therapy , *MYOBLASTS , *PROGENITOR cells , *PLURIPOTENT stem cells - Abstract
Heart failure is the leading cause of death worldwide and the main reason for hospitalization of patients 65-years of age and older. Current therapies only delay progression of the disease but the overall mortality remains as high as 20% per year. The underlying issue is the same whether the cause is acute damage, chronic stress from disease, or aging -- progressive loss of functional cardiomyocytes and diminished hemodynamic output. Trials for regenerating cardiac muscle with cell-based therapy are ongoing for more than 10 years now. Multiple candidate cell types have been used in laboratory experiments and I generation clinical trials, including: skeletal myoblasts, bone marrow and peripheral blood mononuclear cells, endothelial progenitor cells, neonatal cardiomyocytes, mesenchymal stem cells, embryonic stem cells, induced pluripotent stem cells and cardiac stem cells. Results of these studies suggest that cell-based therapy for the failing heart can improve cardiac function. More recently, it has been shown that the heart is not terminally differentiated, and resident cardiac stem cells become a potential source for regeneration of cardiac muscle, smooth muscle and endothelium. This paper summarizes the results of I generation clinical trials and discuss prospects of regenerative therapy in the near future, especially in the context of new stem cell populations available. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
409. Myocardial regeneration by transplantation of modified endothelial progenitor cells expressing SDF-1 in a rat model.
- Author
-
Schuh, Alexander, Kroh, Andreas, Konschalla, Simone, Liehn, Elisa A., Sobota, Radoslav M., Biessen, Erik AL., Bot, Ilze, Tolga Taha, Sönmez, Schober, Andreas, Marx, Nikolaus, Weber, Christian, and Sasse, Alexander
- Subjects
MYOCARDIAL infarction ,ENDOTHELIAL cells ,CELL transplantation ,PROGENITOR cells ,CHEMOKINES ,CARDIAC regeneration ,ECHOCARDIOGRAPHY ,LABORATORY rats - Abstract
Cell based therapy has been shown to attenuate myocardial dysfunction after myocardial infarction (MI) in different acute and chronic animal models. It has been further shown that stromal-cell derived factor-1α ( SDF-1α) facilitates proliferation and migration of endogenous progenitor cells into injured tissue. The aim of the present study was to investigate the role of exogenously applied and endogenously mobilized cells in a regenerative strategy for MI therapy. Lentivirally SDF-1α-infected endothelial progenitor cells ( EPCs) were injected after 90 min. of ligation and reperfusion of the left anterior descending artery ( LAD) intramyocardial and intracoronary using a new rodent catheter system. Eight weeks after transplantation, echocardiography and isolated heart studies revealed a significant improvement of LV function after intramyocardial application of lentiviral with SDF-1 infected EPCs compared to medium control. Intracoronary application of cells did not lead to significant differences compared to medium injected control hearts. Histology showed a significantly elevated rate of apoptotic cells and augmented proliferation after transplantation of EPCs and EPCs + SDF-1α in infarcted myocardium. In addition, a significant increased density of CD31
+ vessel structures, a lower collagen content and higher numbers of inflammatory cells after transplantation of SDF-1 transgenic cells were detectable. Intramyocardial application of lentiviral-infected EPCs is associated with a significant improvement of myocardial function after infarction, in contrast to an intracoronary application. Histological results revealed a significant augmentation of neovascularization, lower collagen content, higher numbers of inflammatory cells and remarkable alterations of apoptotic/proliferative processes in infarcted areas after cell transplantation. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
410. Regenerating functional heart tissue for myocardial repair.
- Author
-
Alcon, Andre, Cagavi Bozkulak, Esra, and Qyang, Yibing
- Subjects
- *
HEART cells , *MYOCARDIAL infarction , *HEART failure , *TISSUE engineering , *HEART transplantation , *CARDIOMYOPLASTY - Abstract
Heart disease is one of the leading causes of death worldwide and the number of patients with the disease is likely to grow with the continual decline in health for most of the developed world. Heart transplantation is one of the only treatment options for heart failure due to an acute myocardial infarction, but limited donor supply and organ rejection limit its widespread use. Cellular cardiomyoplasty, or cellular implantation, combined with various tissue-engineering methods aims to regenerate functional heart tissue. This review highlights the numerous cell sources that have been used to regenerate the heart as well as cover the wide range of tissue-engineering strategies that have been devised to optimize the delivery of these cells. It will probably be a long time before an effective regenerative therapy can make a serious impact at the bedside. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
411. The regenerative capacity of the zebrafish heart is dependent on TGF&bgr; signaling.
- Author
-
Chablais, Fabian and Jaźwińska, Anna
- Subjects
- *
ZEBRA danio , *REGENERATION (Biology) , *CARDIAC regeneration , *CELLULAR signal transduction , *MYOCARDIAL infarction , *SCARS , *FIBROSIS - Abstract
Mammals respond to a myocardial infarction by irreversible scar formation. By contrast, zebrafish are able to resolve the scar and to regenerate functional cardiac muscle. It is not known how opposing cellular responses of fibrosis and new myocardium formation are spatially and temporally coordinated during heart regeneration in zebrafish. Here, we report that the balance between the reparative and regenerative processes is achieved through Smad3-dependent TGF&bgr; signaling. The type I receptor alk5b (tgfbr1b) is expressed in both fibrotic and cardiac cells of the injured heart. TGF&bgr; ligands are locally induced following cryoinjury and activate the signaling pathway both in the infarct area and in cardiomyocytes in the vicinity of the trauma zone. Inhibition of the relevant type I receptors with the specific chemical inhibitor SB431542 qualitatively altered the infarct tissue and completely abolished heart regeneration. We show that transient scar formation is an essential step to maintain robustness of the damaged ventricular wall prior to cardiomyocyte replacement. Taking advantage of the reversible action of the inhibitor, we dissected the multifunctional role of TGF&bgr; signaling into three crucial processes: collagen-rich scar deposition, Tenascin Cassociated tissue remodeling at the infarct-myocardium interface, and cardiomyocyte proliferation. Thus, TGF? signaling orchestrates the beneficial interplay between scar-based repair and cardiomyocyte-based regeneration to achieve complete heart regeneration. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
412. Cardiac telocytes - their junctions and functional implications.
- Author
-
Gherghiceanu, Mihaela and Popescu, Laurentiu
- Subjects
- *
INTERSTITIAL cells , *HEART cells , *PROGENITOR cells , *CELLULAR signal transduction , *CELL communication , *STEM cells , *IMMUNE system , *SCHWANN cells - Abstract
Telocytes (TCs) form a cardiac network of interstitial cells. Our previous studies have shown that TCs are involved in heterocellular contacts with cardiomyocytes and cardiac stem/progenitor cells. In addition, TCs frequently establish 'stromal synapses' with several types of immunoreactive cells in various organs (). Using electron microscopy (EM) and electron microscope tomography (ET), we further investigated the interstitial cell network of TCs and found that TCs form 'atypical' junctions with virtually all types of cells in the human heart. EM and ET showed different junction types connecting TCs in a network ( puncta adhaerentia minima, processus adhaerentes and manubria adhaerentia). The connections between TCs and cardiomyocytes are 'dot' junctions with nanocontacts or asymmetric junctions. Junctions between stem cells and TCs are either 'stromal synapses' or adhaerens junctions. An unexpected finding was that TCs have direct cell-cell (nano)contacts with Schwann cells, endothelial cells and pericytes. Therefore, ultrastructural analysis proved that the cardiac TC network could integrate the overall 'information' from vascular system (endothelial cells and pericytes), nervous system (Schwann cells), immune system (macrophages, mast cells), interstitium (fibroblasts, extracellular matrix), stem cells/progenitors and working cardiomyocytes. Generally, heterocellular contacts occur by means of minute junctions ( point contacts, nanocontacts and planar contacts) and the mean intermembrane distance is within the macromolecular interaction range (10-30 nm). In conclusion, TCs make a network in the myocardial interstitium, which is involved in the long-distance intercellular signaling coordination. This integrated interstitial system appears to be composed of large homotropic zones (TC-TC junctions) and limited (distinct) heterotropic zones (heterocellular junctions of TCs). [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
413. Human spermatagonial stem cells: a novel therapeutic hope for cardiac regeneration and repair?
- Published
- 2012
- Full Text
- View/download PDF
414. Bioengineering Heart Muscle: A Paradigm for Regenerative Medicine.
- Author
-
Vunjak-Novakovic, Gordana, Lui, Kathy O., Tandon, Nina, and Chien, Kenneth R.
- Subjects
- *
ORGANS (Anatomy) , *HEART , *BIOENGINEERING , *REGENERATION (Biology) , *REGENERATIVE medicine , *TISSUE engineering - Abstract
The idea of extending the lifetime of our organs is as old as humankind, fueled by major advances in organ transplantation, novel drugs, and medical devices. However, true regeneration of human tissue has become increasingly plausible only in recent years. The human heart has always been a focus of such efforts, given its notorious inability to repair itself following injury or disease. We discuss here the emerging bioengineering approaches to regeneration of heart muscle as a paradigm for regenerative medicine. Our focus is on biologically inspired strategies for heart regeneration, knowledge gained thus far about how to make a ''perfect'' heart graft, and the challenges that remain to be addressed for tissue-engineered heart regeneration to become a clinical reality. We emphasize the need for interdisciplinary research and training, as recent progress in the field is largely being made at the interfaces between cardiology, stem cell science, and bioengineering. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
415. Cardiac cell therapy: where we've been, where we are, and where we should be headed.
- Author
-
Malliaras, Konstantinos and Marbán, Eduardo
- Subjects
CELLULAR therapy ,HEART cells ,STEM cells ,CARDIOMYOPATHIES ,CARDIAC regeneration ,ISCHEMIA ,ANIMAL models in research ,BONE marrow cells - Abstract
Introduction Stem cell therapy has emerged as a promising strategy for the treatment of ischemic cardiomyopathy. Sources of data Multiple candidate cell types have been used in preclinical animal models and in clinical trials to repair or regenerate the injured heart either directly (through formation of new transplanted tissue) or indirectly (through paracrine effects activating endogenous regeneration). Areas of agreement (i) Clinical trials examining the safety and efficacy of bone marrow derived cells in patients with heart disease are promising, but results leave much room for improvement. (ii) The safety profile has been quite favorable. (iii) Efficacy has been inconsistent and, overall, modest. (iv) Tissue retention of cells after delivery into the heart is disappointingly low. (v) The beneficial effects of adult stem cell therapy are predominantly mediated by indirect paracrine mechanisms. Areas of controversy The cardiogenic potential of bone marrow-derived cells, the mechanism whereby small numbers of poorly-retained cells translate to measurable clinical benefit, and the overall impact on clinical outcomes are hotly debated. Growing points/areas timely for developing research This overview of the field leaves us with cautious optimism, while motivating a search for more effective delivery methods, better strategies to boost cell engraftment, more apt patient populations, safe and effective ‘off the shelf’ cell products and more potent cell types. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
416. Heart of Newt: A Recipe for Regeneration.
- Author
-
Singh, Bhairab, Koyano-Nakagawa, Naoko, Garry, John, and Weaver, Cyprian
- Abstract
The field of regenerative medicine holds tremendous promise for the treatment of chronic diseases. While the adult mammalian heart has limited regenerative capacity, previous studies have focused on cellular therapeutic strategies in an attempt to modulate cardiac regeneration. An alternative strategy relies on the modulation of endogenous stem/progenitor cells or signaling pathways to promote cardiac regeneration. Several organisms, including the newt, have an incomparable capacity for the regeneration of differentiated tissues. An enhanced understanding of the signals, pathways, and factors that mediate the regenerative response in these organisms may be useful in modulating the regenerative response of mammalian organs including the injured adult heart. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
417. Stem Cells in the Infarcted Heart.
- Author
-
Singla, Dinender
- Abstract
Stem cell transplantation is currently generating a significant interest for use in the future treatment of cardiovascular diseases. Stem cell populations are rapidly increasing, and we are still in the search of optimal cell types to use in clinical trials as bone marrow stem cells did not show significant improvement in cardiac function following transplantation. Experimental stem cell studies raised the question on the true differentiation of tissue-specific cell types following transplantation. In fact, recent studies suggest that improved cardiac function is associated with inhibition of apoptosis and fibrosis provided by factors released from stem cells following transplantation. In this review, we will discuss the effects of transplanted stem cells on engraftment and differentiation as well as factors released from stem cells on apoptosis and cardiac remodeling. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
418. Growth plasticity of the embryonic and fetal heart.
- Author
-
Drenckhahn, Jörg-Detlef
- Subjects
- *
FETAL development , *PREVENTION of heart diseases , *FETAL heart , *MORPHOLOGY , *PHYSIOLOGICAL adaptation , *POSTNATAL care - Abstract
The article presents a study that aims to offer a summary of the basic mechanisms of pre- and postnatal cardiac growth as well as the growth plasticity of the embryonic and fetal heart. It notes that various physiological and pathological changes in the intrauterine environment affect the normal cardiac function, tissue composition, and morphology during fetal development. With this, further studies on growth plasticity can help discover ways to prevent heart diseases during postnatal life.
- Published
- 2009
- Full Text
- View/download PDF
419. Cell-Cycle-Based Strategies to Drive Myocardial Repair.
- Author
-
Zhu, Wuqiang, Hassink, Rutger J., Rubart, Michael, and Field, Loren J.
- Subjects
- *
HEART cells , *CELL proliferation , *STEM cell research , *HEMODYNAMICS , *ARTERIAL occlusions , *PHYSIOLOGY - Abstract
Cardiomyocytes exhibit robust proliferative activity during development. After birth, cardiomyocyte proliferation is markedly reduced. Consequently, regenerative growth in the postnatal heart via cardiomyocyte proliferation (and, by inference, proliferation of stem-cell-derived cardiomyocytes) is limited and often insufficient to affect repair following injury. Here, we review studies wherein cardiomyocyte cell cycle proliferation was induced via targeted expression of cyclin D2 in postnatal hearts. Cyclin D2 expression resulted in a greater than 500-fold increase in cell cycle activity in transgenic mice as compared to their nontransgenic siblings. Induced cell cycle activity resulted in infarct regression and concomitant improvement in cardiac hemodynamics following coronary artery occlusion. These studies support the notion that cell-cycle-based strategies can be exploited to drive myocardial repair following injury. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
420. From Spheroids to Organoids: The Next Generation of Model Systems of Human Cardiac Regeneration in a Dish.
- Author
-
Scalise, Mariangela, Marino, Fabiola, Salerno, Luca, Cianflone, Eleonora, Molinaro, Claudia, Salerno, Nadia, De Angelis, Antonella, Viglietto, Giuseppe, Urbanek, Konrad, and Torella, Daniele
- Subjects
- *
CARDIAC regeneration , *BIOLOGICAL evolution , *PLURIPOTENT stem cells , *BIOLOGICAL systems , *ORGANOIDS - Abstract
Organoids are tiny, self-organized, three-dimensional tissue cultures that are derived from the differentiation of stem cells. The growing interest in the use of organoids arises from their ability to mimic the biology and physiology of specific tissue structures in vitro. Organoids indeed represent promising systems for the in vitro modeling of tissue morphogenesis and organogenesis, regenerative medicine and tissue engineering, drug therapy testing, toxicology screening, and disease modeling. Although 2D cell cultures have been used for more than 50 years, even for their simplicity and low-cost maintenance, recent years have witnessed a steep rise in the availability of organoid model systems. Exploiting the ability of cells to re-aggregate and reconstruct the original architecture of an organ makes it possible to overcome many limitations of 2D cell culture systems. In vitro replication of the cellular micro-environment of a specific tissue leads to reproducing the molecular, biochemical, and biomechanical mechanisms that directly influence cell behavior and fate within that specific tissue. Lineage-specific self-organizing organoids have now been generated for many organs. Currently, growing cardiac organoid (cardioids) from pluripotent stem cells and cardiac stem/progenitor cells remains an open challenge due to the complexity of the spreading, differentiation, and migration of cardiac muscle and vascular layers. Here, we summarize the evolution of biological model systems from the generation of 2D spheroids to 3D organoids by focusing on the generation of cardioids based on the currently available laboratory technologies and outline their high potential for cardiovascular research. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
421. Loss of m6A methyltransferase METTL3 promotes heart regeneration and repair after myocardial injury.
- Author
-
Gong, Rui, Wang, Xiuxiu, Li, Hanjing, Liu, Shenzhen, Jiang, Zuke, Zhao, Yiming, Yu, Yang, Han, Zhenbo, Yu, Ying, Dong, Chaorun, Li, Shuainan, Xu, Binbin, Zhang, Wenwen, Wang, Ning, Li, Xingda, Gao, Xinlu, Yang, Fan, Bamba, Djibril, Ma, Wenya, and Liu, Yu
- Subjects
- *
MYOCARDIAL injury , *CARDIAC regeneration , *MESSENGER RNA , *KNOCKOUT mice , *MICRORNA , *HEART - Abstract
N6-Methyladenosine (m6A), one of the important epigenitic modifications, is very commom in messenger RNAs (mRNAs) of eukaryotes, and has been involved in various diseases. However, the role of m6A modification in heart regeneration after injury remains unclear. The study was conducted to investigate whether targeting methyltransferase-like 3 (METTL3) could replenish the loss of cardiomyocytes (CMs) and improve cardiac function after myocardial infarction (MI). METTL3 knockout mouse line was generated. A series of functional experiments were carried out and the molecular mechanism was further explored. We identified that METTL3, a methyltransferase of m6A methylation, is upregulated in mouse hearts after birth, which is the opposite of the changes in CMs proliferation. Furthermore, both METTL3 heterozygous knockout mice and administration of METTL3 shRNA adenovirus in mice exhibited CMs cell cycle re-entered, infract size decreased and cardiac function improved after MI. Mechanically, the silencing of METTL3 promoted CMs proliferation by reducing primary miR-143 (pri-miR-143) m6A modificaiton, thereby inhibiting the pri-miR-143 into mature miR‐143‐3p. Moreover, we found that miR-143–3p has targeting effects on Yap and Ctnnd1 so as to regulate CMs proliferation. METTL3 deficiency contributes to heart regeneration after MI via METTL3-pri-miR-143-(miR-143)-Yap/Ctnnd1 axis. This study provides new insights into the significance of RNA m6A modification in heart regeneration. [Display omitted] [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
422. Common marmoset embryonic stem cell can differentiate into cardiomyocytes
- Author
-
Chen, Hao, Hattori, Fumiyuki, Murata, Mitsushige, Li, Weizhen, Yuasa, Shinsuke, Onizuka, Takeshi, Shimoji, Kenichiro, Ohno, Yohei, Sasaki, Erika, Kimura, Kensuke, Hakuno, Daihiko, Sano, Motoaki, Makino, Shinji, Ogawa, Satoshi, and Fukuda, Keiichi
- Subjects
- *
EMBRYONIC stem cells , *HEART cells , *STEM cells , *MONKEYS - Abstract
Abstract: Common marmoset monkeys have recently attracted much attention as a primate research model, and are preferred to rhesus and cynomolgus monkeys due to their small bodies, easy handling and efficient breeding. We recently reported the establishment of common marmoset embryonic stem cell (CMESC) lines that could differentiate into three germ layers. Here, we report that our CMESC can also differentiate into cardiomyocytes and investigated their characteristics. After induction, FOG-2 was expressed, followed by GATA4 and Tbx20, then Nkx2.5 and Tbx5. Spontaneous beating could be detected at days 12–15. Immunofluorescent staining and ultrastructural analyses revealed that they possessed characteristics typical of functional cardiomyocytes. They showed sinus node-like action potentials, and the beating rate was augmented by isoproterenol stimulation. The BrdU incorporation assay revealed that CMESC-derived cardiomyocytes retained a high proliferative potential for up to 24 weeks. We believe that CMESC-derived cardiomyocytes will advance preclinical studies in cardiovascular regenerative medicine. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
423. Plasmid-mediated VEGF gene transfer induces cardiomyogenesis and reduces myocardial infarct size in sheep.
- Author
-
Vera Janavel, G., Crottogini, A., Cabeza Meckert, P., Cuniberti, L., Mele, A., Papouchado, M., Fernández, N., Bercovich, A., Criscuolo, M., Melo, C., and Laguens, R.
- Subjects
- *
VASCULAR endothelial growth factors , *GENETIC transformation , *MYOCARDIAL infarction , *CORONARY disease , *CYTOKINESIS , *HEART cells , *SHEEP - Abstract
We have recently reported that in pigs with chronic myocardial ischemia heart transfection with a plasmid encoding the 165 isoform of human vascular endothelial growth factor (pVEGF165) induces an increase in the mitotic index of adult cardiomyocytes and cardiomyocyte hyperplasia. On these bases we hypothesized that VEGF gene transfer could also modify the evolution of experimental myocardial infarct. In adult sheep pVEGF165 (3.8 mg, n=7) or empty plasmid (n=7) was injected intramyocardially 1 h after coronary artery ligation. After 15 days infarct area was 11.3±1.3% of the left ventricle in the VEGF group and 18.2±2.1% in the empty plasmid group (P<0.02). The mechanisms involved in infarct size reduction (assessed in additional sheep at 7 and 10 days after infarction) included an increase in early angiogenesis and arteriogenesis, a decrease in peri-infarct fibrosis, a decrease in myofibroblast proliferation, enhanced cardiomyoblast proliferation and mitosis of adult cardiomyocytes with occasional cytokinesis. Resting myocardial perfusion (99mTc-sestamibi SPECT) was higher in VEGF-treated group than in empty plasmid group 15 days after myocardial infarction. We conclude that plasmid-mediated VEGF gene transfer reduces myocardial infarct size by a combination of effects including neovascular proliferation, modification of fibrosis and cardiomyocyte regeneration.Gene Therapy (2006) 13, 1133–1142. doi:10.1038/sj.gt.3302708; published online 6 April 2006 [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
424. Molecular barriers to direct cardiac reprogramming
- Author
-
Jiandong Liu, Haley Ruth Vaseghi, and Li Qian
- Subjects
0301 basic medicine ,Cardiac function curve ,heart regeneration ,Intracellular Space ,lcsh:Animal biochemistry ,Review ,Cell fate determination ,Biology ,Biochemistry ,cardiac reprogramming ,Epigenesis, Genetic ,03 medical and health sciences ,Drug Discovery ,microRNA ,medicine ,Animals ,Humans ,Epigenetics ,Myocardial infarction ,lcsh:QH573-671 ,Transcription factor ,lcsh:QP501-801 ,epigenetics ,business.industry ,lcsh:Cytology ,Myocardium ,Cell Biology ,Cellular Reprogramming ,medicine.disease ,3. Good health ,Biotechnology ,Cell biology ,030104 developmental biology ,myocardial infarction ,Intercellular Signaling Peptides and Proteins ,Stem cell ,business ,Reprogramming ,Signal Transduction - Abstract
Myocardial infarction afflicts close to three quarters of a million Americans annually, resulting in reduced heart function, arrhythmia, and frequently death. Cardiomyocyte death reduces the heart’s pump capacity while the deposition of a non-conductive scar incurs the risk of arrhythmia. Direct cardiac reprogramming emerged as a novel technology to simultaneously reduce scar tissue and generate new cardiomyocytes to restore cardiac function. This technology converts endogenous cardiac fibroblasts directly into induced cardiomyocyte-like cells using a variety of cocktails including transcription factors, microRNAs, and small molecules. Although promising, direct cardiac reprogramming is still in its fledging phase, and numerous barriers have to be overcome prior to its clinical application. This review discusses current findings to optimize reprogramming efficiency, including reprogramming factor cocktails and stoichiometry, epigenetic barriers to cell fate reprogramming, incomplete conversion and residual fibroblast identity, requisite growth factors, and environmental cues. Finally, we address the current challenges and future directions for the field.
- Published
- 2017
425. Notch Inhibition Enhances Cardiac Reprogramming by Increasing MEF2C Transcriptional Activity
- Author
-
Rhonda Bassel-Duby, Huanyu Zhou, María Abad, María Gabriela Morales, Beibei Chen, Eric N. Olson, and Hisayuki Hashimoto
- Subjects
0301 basic medicine ,transdifferentiation ,Cellular differentiation ,cardiomyocytes ,Biochemistry ,Mice ,Myocyte ,MEF2C ,Myocytes, Cardiac ,lcsh:QH301-705.5 ,Notch signaling ,Genetics ,lcsh:R5-920 ,Receptors, Notch ,MEF2 Transcription Factors ,Transdifferentiation ,cell-fate conversion ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Cellular Reprogramming ,Cell biology ,DAPT ,lcsh:Medicine (General) ,Reprogramming ,Signal Transduction ,Sarcomeres ,Transcriptional Activation ,animal structures ,heart regeneration ,direct cellular reprogramming ,Notch signaling pathway ,regenerative medicine ,Biology ,Diamines ,Article ,03 medical and health sciences ,Calcium flux ,Animals ,Calcium Signaling ,Transcription factor ,Gene Expression Profiling ,Cell Biology ,Electrophysiological Phenomena ,Thiazoles ,030104 developmental biology ,lcsh:Biology (General) ,Transcriptome ,Proto-Oncogene Proteins c-akt ,Developmental Biology - Abstract
Summary Conversion of fibroblasts into functional cardiomyocytes represents a potential means of restoring cardiac function after myocardial infarction, but so far this process remains inefficient and little is known about its molecular mechanisms. Here we show that DAPT, a classical Notch inhibitor, enhances the conversion of mouse fibroblasts into induced cardiac-like myocytes by the transcription factors GATA4, HAND2, MEF2C, and TBX5. DAPT cooperates with AKT kinase to further augment this process, resulting in up to 70% conversion efficiency. Moreover, DAPT promotes the acquisition of specific cardiomyocyte features, substantially increasing calcium flux, sarcomere structure, and the number of spontaneously beating cells. Transcriptome analysis shows that DAPT induces genetic programs related to muscle development, differentiation, and excitation-contraction coupling. Mechanistically, DAPT increases binding of the transcription factor MEF2C to the promoter regions of cardiac structural genes. These findings provide mechanistic insights into the reprogramming process and may have important implications for cardiac regeneration therapies., Graphical Abstract, Highlights • Notch activation is a barrier for GHMT-induced cardiac cell reprogramming • Notch blockade by DAPT improves GHMT-induced cardiac reprogramming • DAPT increases sarcomere organization, calcium flux, and beating in GHMT reprogramming • DAPT enhances transcriptional activity of MEF2C in GHMT reprogramming, In this article, Olson and colleagues show that Notch signaling activation is a critical barrier for cardiac cell reprogramming. Notch signaling blockade by DAPT, a γ-secretase inhibitor, enhances fibroblast conversion to cardiomyocytes by increasing calcium flux, sarcomere structure, and beating. Mechanistically, Notch inhibition enhances the transcriptional activity of the cardiogenic transcription factor MEF2C, thereby enhancing expression of cardiac genes.
- Published
- 2017
426. Stem Cell Technology in Cardiac Regeneration: A Pluripotent Stem Cell Promise
- Author
-
Maurilio Sampaolesi and Robin Duelen
- Subjects
0301 basic medicine ,Pluripotent Stem Cells ,Pathology ,medicine.medical_specialty ,Cellular differentiation ,Population ,lcsh:Medicine ,Review ,Biology ,Bioinformatics ,Regenerative Medicine ,Regenerative medicine ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Heart disorder ,Stem cell-derived exosome ,medicine ,Animals ,Humans ,Regeneration ,Myocytes, Cardiac ,Human pluripotent stem cell ,education ,Induced pluripotent stem cell ,Heart Failure ,education.field_of_study ,lcsh:R5-920 ,Embryonic cardiomyogenesis ,Regeneration (biology) ,lcsh:R ,Cell Differentiation ,General Medicine ,embryonic cardiomyogenesis ,heart regeneration ,human pluripotent stem cell ,stem cell-based therapy ,stem cell-derived exosome ,animals ,heart failure ,humans ,models ,biological ,myocytes ,cardiac ,pluripotent stem cells ,regeneration ,regenerative medicine ,treatment outcome ,cell differentiation ,medicine (all) ,biochemistry ,genetics and molecular biology (all) ,030104 developmental biology ,Treatment Outcome ,Stem cell ,Stem cell-based therapy ,lcsh:Medicine (General) ,Heart regeneration ,Adult stem cell - Abstract
Despite advances in cardiovascular biology and medical therapy, heart disorders are the leading cause of death worldwide. Cell-based regenerative therapies become a promising treatment for patients affected by heart failure, but also underline the need for reproducible results in preclinical and clinical studies for safety and efficacy. Enthusiasm has been tempered by poor engraftment, survival and differentiation of the injected adult stem cells. The crucial challenge is identification and selection of the most suitable stem cell type for cardiac regenerative medicine. Human pluripotent stem cells (PSCs) have emerged as attractive cell source to obtain cardiomyocytes (CMs), with potential applications, including drug discovery and toxicity screening, disease modelling and innovative cell therapies. Lessons from embryology offered important insights into the development of stem cell-derived CMs. However, the generation of a CM population, uniform in cardiac subtype, adult maturation and functional properties, is highly recommended. Moreover, hurdles regarding tumorigenesis, graft cell death, immune rejection and arrhythmogenesis need to be overcome in clinical practice. Here we highlight the recent progression in PSC technologies for the regeneration of injured heart. We review novel strategies that might overcome current obstacles in heart regenerative medicine, aiming at improving cell survival and functional integration after cell transplantation., Highlights • Human pluripotent stem cells emerge as attractive tool for cardiac regeneration approaches. • Plasticity of human pluripotent stem cells towards cardiac-related cell types guarantees repopulation of injured heart. • Combination of stem cell and gene editing therapies has potential to become next generation treatment for cardiac diseases. Data for this Review were identified by searches of MEDLINE and PubMed, and references from relevant articles using the search terms “cardiomyogenesis”, “adult stem cells”, “pluripotent stem cells” and “cardiac regeneration”. Only articles published in English between 1976 and 2017 were included. The majority of the articles reported were published after 2000.
- Published
- 2017
427. Enhancement by growth factors of cardiac myocyte differentiation from embryonic stem cells: A promising foundation for cardiac regeneration
- Author
-
Singla, Dinender K. and Sobel, Burton E.
- Subjects
- *
GROWTH factors , *EMBRYONIC stem cells , *CELL transplantation , *HEART diseases - Abstract
Abstract: Cell transplantation is a promising, still novel, potentially therapeutic approach for the treatment of heart diseases. Clinical applications require generation of large number of donor cells. Embryonic stem (ES) cells are capable of self-renewal apparently in an unlimited fashion, in vitro. Theoretically, they can differentiate into any cell type required for cell transplantation, including cardiac myocytes. Diverse growth factors have been implicated in programming diverse cellular processes, including development of the embryonic heart, ES cell self-renewal, and cardiac myocyte differentiation from ES cells. This review addresses the current understanding of the role of growth factors in the differentiation of cardiac myocytes from ES–embryoid body cell systems in vitro as well as cardiac regeneration in vivo. [Copyright &y& Elsevier]
- Published
- 2005
- Full Text
- View/download PDF
428. Recent advances in myocardial regeneration strategy
- Author
-
Bingren Gao, Kai Sheng, and Yu Nie
- Subjects
0301 basic medicine ,Medicine (General) ,Heart Diseases ,proliferation ,Review ,030204 cardiovascular system & hematology ,Biochemistry ,cardiomyocyte injury ,03 medical and health sciences ,0302 clinical medicine ,Animal model ,R5-920 ,Animals ,Humans ,Regeneration ,Medicine ,Myocytes, Cardiac ,Cell Proliferation ,business.industry ,animal model ,Regeneration (biology) ,Biochemistry (medical) ,Heart ,Cell Biology ,General Medicine ,Cardiovascular physiology ,030104 developmental biology ,repair ,business ,Neuroscience ,Heart regeneration - Abstract
This review described the current status of research into the regeneration potential of myocardial cells after myocardial injury, focussing on possible mechanisms of regeneration and the application of animal models to human biology, all with the aim of evaluating any novel approaches to the regeneration of human cardiomyocytes. A literature review was undertaken of the PubMed® and The Cochrane Library databases using the search terms ‘regeneration’, ‘heart regeneration’, ‘cardiac regeneration’, ‘proliferation’, ‘animal model’, ‘repair’ and ‘myocardial cell injury’ in English language publications only. The search covered publications between 1 January 2002 to 31 December 2017. The cardiac regeneration capability significantly differed among different species. In lower vertebrates, such as zebrafish, cardiomyocytes possess a sustained regeneration capacity under specific conditions. In mammalian animals, such as mice, the cardiomyocytes retain a regeneration capability under specific conditions, which gradually declines. Inflammation, non-coding RNA, gene regulatory elements, signal transduction and cell phenotype transformation play pivotal roles in cardiomyocyte regeneration. Myocardial regeneration appears to be a viable repair strategy for cardiomyocyte loss, which deserves further research in order to validate its clinical applicability in humans.
- Published
- 2019
429. Cardiac Reprogramming Factors Synergistically Activate Genome-wide Cardiogenic Stage-Specific Enhancers
- Author
-
Axel Visel, Eric N. Olson, Wenduo Ye, Glynnis A. Garry, Giovanni A. Botten, Ning Liu, Hisayuki Hashimoto, Rhonda Bassel-Duby, Diane E. Dickel, Zhaoning Wang, Marco Osterwalder, Huanyu Zhou, and Venkat S. Malladi
- Subjects
Gata4 ,Gene regulatory network ,cardiomyocytes ,Inbred C57BL ,Cardiovascular ,Medical and Health Sciences ,Mice ,0302 clinical medicine ,Basic Helix-Loop-Helix Transcription Factors ,MEF2C ,Myocytes, Cardiac ,Gene Regulatory Networks ,Cells, Cultured ,0303 health sciences ,Cultured ,GATA4 ,MEF2 Transcription Factors ,Biological Sciences ,Cellular Reprogramming ,Tbx5 ,Cell biology ,ErbB Receptors ,Heart Disease ,embryonic structures ,Molecular Medicine ,HAND2 ,Reprogramming ,Cardiac ,Signal Transduction ,heart regeneration ,Cells ,Hand2 ,Biology ,Article ,03 medical and health sciences ,Genetics ,Animals ,Epigenetics ,Enhancer ,Transcription factor ,030304 developmental biology ,Myocytes ,Akt1 ,induced cardiac-like myocytes ,Human Genome ,Cell Biology ,Fibroblasts ,Mef2c ,direct reprogramming ,GATA4 Transcription Factor ,Mice, Inbred C57BL ,biology.protein ,T-Box Domain Proteins ,030217 neurology & neurosurgery ,Genome-Wide Association Study ,Developmental Biology - 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 sitesand 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.
- Published
- 2019
430. Proteomics analysis of extracellular matrix remodeling during zebrafish heart regeneration
- Author
-
Daniel Navajas, Anna Garcia-Puig, Cristina García-Pastor, Angel Raya, Senda Jiménez-Delgado, Ignasi Jorba, Francesc Canals, and Jose Luis Mosquera
- Subjects
Proteomics ,heart regeneration ,Microscopy, Atomic Force ,Biochemistry ,Analytical Chemistry ,Extracellular matrix ,03 medical and health sciences ,Response to injury ,Developmental biology ,Animals ,Regeneration ,Cytoskeleton ,Molecular Biology ,Zebrafish ,030304 developmental biology ,0303 health sciences ,Extracellular Matrix Proteins ,atomic force microscopy ,Decellularization ,biology ,Malalties cardiovasculars ,Atomic force microscopy ,Myocardium ,Research ,Regeneration (biology) ,030302 biochemistry & molecular biology ,Heart ,Animal models in research ,Zebrafish Proteins ,Cardiovascular disease ,proteomic analysis ,biology.organism_classification ,Biomechanical Phenomena ,Animal models ,Cell biology ,Cardiovascular diseases ,Cardiovascular function or biology ,Biologia del desenvolupament ,Models animals en la investigació - Abstract
Zebrafish can regenerate their hearts. The role of the extracellular matrix in this process is largely unknown. We have analyzed the proteome in control hearts and at different times of regeneration. Decellularization of samples allowed for enrichment of extracellular matrix proteins, increasing their detection. The results reported dynamic changes in specific proteins associated with specific stages of the regenerative process. Biomechanical analysis by atomic force microscopy revealed concomitant changes in matrix stiffness during this process., Graphical Abstract Highlights We have developed a decellularization protocol for ECM protein enrichment. We have characterized the proteome of adult zebrafish heart ECM. We describe dynamic changes in heart ECM proteome during regeneration. We describe changes in heart ECM stiffness during regeneration., 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.
- Published
- 2019
431. Concise Review: Reduction of Adverse Cardiac Scarring Facilitates Pluripotent Stem Cell-Based Therapy for Myocardial Infarction
- Author
-
Xiangnan Li, Jialiang Liang, Lin Jiang, Yigang Wang, Christian Paul, and Wei Huang
- Subjects
0301 basic medicine ,Pluripotent Stem Cells ,Cardiac fibrosis ,Cell ,Myocardial Infarction ,Bioengineering ,Biology ,Regenerative Medicine ,Mechanotransduction, Cellular ,Cell therapy ,03 medical and health sciences ,Paracrine signalling ,Cicatrix ,0302 clinical medicine ,Fibrosis ,Paracrine Communication ,medicine ,Animals ,Humans ,Regeneration ,Myocytes, Cardiac ,Induced pluripotent stem cell ,Myofibroblasts ,Cardiomyocytes ,Tissue Engineering ,Graft Survival ,Hydrogels ,Cell Biology ,Genetic Therapy ,medicine.disease ,Disease Models, Animal ,MicroRNAs ,Tissue stiffness ,030104 developmental biology ,medicine.anatomical_structure ,Cancer research ,Molecular Medicine ,Stem cell ,Hydroxymethylglutaryl-CoA Reductase Inhibitors ,Myofibroblast ,030217 neurology & neurosurgery ,Heart regeneration ,Developmental Biology ,Stem Cell Transplantation - 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.
- Published
- 2019
432. Wilms Tumor 1b Expression Defines a Pro-regenerative Macrophage Subtype and Is Required for Organ Regeneration in the Zebrafish
- Author
-
Steffi Manig, Thomas J. D. Bates, Andrés Sanz-Morejón, Juan Manuel González-Rosa, Christoph Englert, Marta Ruiz-Ortega, Ines J. Marques, Andreas Große, María Galardi-Castilla, Ana M. Briones, Indre Piragyte, Mercedes Salaices, Alexander Ernst, Marius-Alexandru Botos, Xavier Langa, Hanna Reuter, Nadia Mercader, Ana B. García-Redondo, Centro de Investigación Biomedica en Red - CIBER, European Molecular Biology Organization, Swiss National Science Foundation, Unión Europea. Comisión Europea, European Research Council, Instituto de Salud Carlos III, Ministerio de Ciencia, Innovación y Universidades (España), and Fundación ProCNIC
- Subjects
0301 basic medicine ,heart regeneration ,Mutant ,610 Medicine & health ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Fin regeneration ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,medicine ,Animals ,Regeneration ,Macrophage ,WT1 Proteins ,Zebrafish ,lcsh:QH301-705.5 ,Organ regeneration ,Innate immune system ,Macrophages ,fin regeneration ,Heart ,Wilms' tumor ,Zebrafish Proteins ,zebrafish ,medicine.disease ,biology.organism_classification ,Cell biology ,030104 developmental biology ,lcsh:Biology (General) ,Wilms tumor 1b ,Animal Fins ,570 Life sciences ,biology ,030217 neurology & neurosurgery ,Heart regeneration - Abstract
Summary Organ regeneration is preceded by the recruitment of innate immune cells, which play an active role during repair and regrowth. Here, we studied macrophage subtypes during organ regeneration in the zebrafish, an animal model with a high regenerative capacity. We identified a macrophage subpopulation expressing Wilms tumor 1b (wt1b), which accumulates within regenerating tissues. This wt1b+ macrophage population exhibited an overall pro-regenerative gene expression profile and different migratory behavior compared to the remainder of the macrophages. Functional studies showed that wt1b regulates macrophage migration and retention at the injury area. Furthermore, wt1b-null mutant zebrafish presented signs of impaired macrophage differentiation, delayed fin growth upon caudal fin amputation, and reduced cardiomyocyte proliferation following cardiac injury that correlated with altered macrophage recruitment to the regenerating areas. We describe a pro-regenerative macrophage subtype in the zebrafish and a role for wt1b in organ regeneration., Graphical Abstract, Highlights • Wt1b+ macrophages reveal a pro-regenerative gene expression prolife • Wt1b controls migration behavior of macrophages during fin and heart regeneration • Wt1b regulates differentiation of macrophages in the kidney marrow • wt1b mutants reveal impaired fin and heart regeneration, Sanz-Morejón et al. identify Wilms tumor 1b (Wt1b)+ macrophages with a pro-regenerative gene signature in injured fins and hearts in the zebrafish. They show that Wt1b controls macrophage migration and differentiation. Regeneration is impaired in wt1b mutants, supporting a role for this gene, likely within macrophages, in organ regeneration.
- Published
- 2019
- Full Text
- View/download PDF
433. Adult sox10+ Cardiomyocytes Contribute to Myocardial Regeneration in the Zebrafish
- Author
-
Sande-Melón, Marcos, Marques, Inês J., Galardi-Castilla, María, Langa, Xavier, Pérez-López, María, Botos, Marius-Alexandru, Sánchez-Iranzo, Héctor, Guzmán-Martínez, Gabriela, Ferreira Francisco, David Miguel, Pavlinic, Dinko, Benes, Vladimir, Bruggmann, Rémy, Mercader Huber, Nadia Isabel, Ministerio de Economía y Competitividad (España), Swiss National Science Foundation, Unión Europea. Comisión Europea, European Research Council, Unión Europea. Fondo Europeo de Desarrollo Regional (FEDER/ERDF), Instituto de Salud Carlos III, Fundación ProCNIC, and Ministerio de Ciencia, Innovación y Universidades (España)
- Subjects
heart regeneration ,sox10 ,SOXE Transcription Factors ,Sox10 ,610 Medicine & health ,Heart ,Zebrafish Proteins ,zebrafish ,Article ,lcsh:Biology (General) ,cardiomyocyte proliferation ,embryonic structures ,Cardiomyocyte proliferation ,Animals ,Regeneration ,570 Life sciences ,biology ,Myocytes, Cardiac ,lcsh:QH301-705.5 ,Cells, Cultured ,Heart regeneration ,Zebrafish ,Cell Proliferation - Abstract
Summary 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., Graphical Abstract, Highlights • Adult sox10-derived cardiomyocytes contribute to zebrafish heart regeneration • sox10-derived cardiomyocytes have a high proliferation index • sox10-derived cardiomyocytes have a distinct gene expression profile • Genetic ablation of sox10-derived cells impairs heart regeneration, Unlike adult mammals, zebrafish regenerate their heart after injury through proliferation of preexistent cardiomyocytes. Sande-Melón et al. identify a subset of sox10-positive cardiomyocytes within the uninjured heart with a high capacity to contribute to the new myocardium. Ablation of these cardiomyocytes confirms that they play an essential role during heart regeneration.
- Published
- 2019
- Full Text
- View/download PDF
434. Chapter 11: Regenerative Medicine and Biomarkers for Dilated Cardiomyopathy
- Author
-
Lesizza, Pierluigi, Aleksova, Aneta, Ortis, Benedetta, Antonio Paolo Beltrami, Giacca, Mauro, Sinagra, Gianfranco, Sinagra, Gianfranco, Merlo, Marco, Pinamonti, Bruno, Lesizza, Pierluigi, Aleksova, Aneta, Ortis, Benedetta, Paolo Beltrami, Antonio, and Giacca, Mauro
- Subjects
Gene therapy ,Heart regeneration ,Cell therapy ,Biomarkers ,Heart failure ,Inflammasome ,Biomarker - 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.
- Published
- 2019
435. Reactivating endogenous mechanisms of cardiac regeneration via paracrine boosting using the human amniotic fluid stem cell secretome
- Author
-
Francesca Campagnoli, Pierangela De Biasio, Agnese Palmeri, Marie-José Goumans, Vittorio Rosti, Ambra Costa, Silvia Moimas, Tessa van Herwaarden, Anke M. Smits, Francesco Santini, Sveva Bollini, Lucio Barile, Kirsten Lodder, Mauro Giacca, Carolina Balbi, Francesco Moccia, University of Zurich, Bollini, Sveva, Balbi, C., Lodder, K., Costa, A., Moimas, S., Moccia, F., van Herwaarden, T., Rosti, V., Campagnoli, F., Palmeri, A., De Biasio, P., Santini, F., Giacca, M., Goumans, M. -J., Barile, L., Smits, A. M., and Bollini, S.
- Subjects
Cardiac function curve ,Amniotic fluid ,heart regeneration ,Angiogenesis ,Cellular differentiation ,Endogeny ,610 Medicine & health ,030204 cardiovascular system & hematology ,11171 Cardiocentro Ticino ,2705 Cardiology and Cardiovascular Medicine ,03 medical and health sciences ,Paracrine signalling ,Mice ,0302 clinical medicine ,stem cells ,Paracrine Communication ,Medicine ,Myocyte ,Animals ,Humans ,Myocytes, Cardiac ,030212 general & internal medicine ,Cells, Cultured ,Heart Failure ,Microscopy, Confocal ,business.industry ,amniotic fluid ,Cell Differentiation ,Cell biology ,Rats ,cardio protection ,stem cell ,Mice, Inbred C57BL ,Disease Models, Animal ,Animals, Newborn ,Culture Media, Conditioned ,Stem cell ,business ,Cardiology and Cardiovascular Medicine ,Stem Cell Transplantation - Abstract
Background: The adult mammalian heart retains residual regenerative capability via endogenous cardiac progenitor cell (CPC) activation and cardiomyocyte proliferation. We previously reported the paracrine cardioprotective capacity of human amniotic fluid-derived stem cells (hAFS) following ischemia or cardiotoxicity. Here we analyse the potential of hAFS secretome fractions for cardiac regeneration and future clinical translation.Methods: hAFS were isolated from amniotic fluid leftover samples from prenatal screening. hAFS conditioned medium (hAFS-CM) was obtained following hypoxic preconditioning. Anti-apoptotic, angiogenic and proliferative effects were evaluated on rodent neonatal cardiomyocytes (r/mNVCM), human endothelial colony forming cells (hECFC) and human CPC. Mice undergoing myocardial infarction (MI) were treated with hAFS-CM, hAFS-extracellular vesicles (hAFS-EV), or EV-depleted hAFS-CM(hAFS-DM) by single intra-myocardial administration and evaluated in the short and long term.Results: hAFS-CM improved mNVCM survival under oxidative and hypoxic damage, induced Ca2+-dependent angiogenesis in hECFC and triggered hCPC and rNVCM proliferation. hAFS-CM treatment after MI counteracted scarring, supported cardiac function, angiogenesis and cardiomyocyte cell cycle progression in the long term. hAFS-DM had no effect. hAFS-CM and hAFS-EV equally induced epicardium WT1+CPC reactivation. Although no CPC cardiovascular differentiation was observed, our data suggests contribution to local angiogenesis by paracrine modulation. hAFS-EV alone were able to recapitulate all the beneficial effects exerted by hAFS-CM, except for stimulation of vessel formation.Conclusions: hAFS-CM and hAFS-EV can improve cardiac repair and trigger cardiac regeneration via paracrine modulation of endogenous mechanisms. While both formulations are effective in sustaining myocardial renewal, hAFS-CM retains higher pro-angiogenic potential, while hAFS-EV particularly enhances cardiac function. (c) 2019 Elsevier B.V. All rights reserved.
- Published
- 2019
436. Heart regeneration, stem cells, and cytokines
- Author
-
Li Na, Wang Chuan, Jia LiXin, and Du Jie
- Subjects
Cytokines ,Heart regeneration ,Stem cells ,Paracrine mechanisms ,Medicine - 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
- Full Text
- View/download PDF
437. Covering and Re-Covering the Heart: Development and Regeneration of the Epicardium
- Author
-
Jingli Cao and Yingxi Cao
- Subjects
0301 basic medicine ,Cardiac function curve ,medicine.medical_specialty ,lcsh:Diseases of the circulatory (Cardiovascular) system ,heart regeneration ,Review ,Injury response ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Pharmacology (medical) ,epicardium ,Myocardial infarction ,General Pharmacology, Toxicology and Pharmaceutics ,Progenitor cell ,Zebrafish ,biology ,Heart development ,business.industry ,proepicardial organ ,Regeneration (biology) ,heart development ,biology.organism_classification ,medicine.disease ,zebrafish ,030104 developmental biology ,lcsh:RC666-701 ,Cardiac repair ,Cardiology ,cardiovascular system ,business ,030217 neurology & neurosurgery - 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
438. Paracrine mechanism of redox signalling for post-mitotic cell and tissue regeneration
- Author
-
Francesco De Virgiliis, Simone Di Giovanni, Celio X.C. Santos, Ajay M. Shah, and Arnau Hervera
- Subjects
GROWTH-FACTOR ,RETINAL GANGLION-CELLS ,NF-KAPPA-B ,DEPENDENT ACTIVATION ,Mitosis ,Biology ,Mitochondrion ,Extracellular Vesicles ,03 medical and health sciences ,Paracrine signalling ,0302 clinical medicine ,NADPH OXIDASE ACTIVITY ,Paracrine Communication ,Animals ,Humans ,Myocytes, Cardiac ,HYPOXIA-INDUCIBLE FACTOR-1-ALPHA ,OXIDATIVE STRESS ,Transcription factor ,11 Medical and Health Sciences ,030304 developmental biology ,Neurons ,AXONAL REGENERATION ,0303 health sciences ,Science & Technology ,Kinase ,Mechanism (biology) ,Regeneration (biology) ,MITOCHONDRIAL COMPLEX-III ,Cell Biology ,06 Biological Sciences ,Cell biology ,Oxidation-Reduction ,Developmental biology ,Life Sciences & Biomedicine ,030217 neurology & neurosurgery ,Intracellular ,Signal Transduction ,HEART REGENERATION ,Developmental Biology - Abstract
Adult postmitotic mammalian cells, including neurons and cardiomyocytes, have a limited capacity to regenerate after injury. Therefore, an understanding of the molecular mechanisms underlying their regenerative ability is critical to advance tissue repair therapies. Recent studies highlight how redox signalling via paracrine cell-to-cell communication may act as a central mechanism coupling tissue injury with regeneration. Post-injury redox paracrine signalling can act by diffusion to nearby cells, through mitochondria or within extracellular vesicles, affecting specific intracellular targets such as kinases, phosphatases, and transcription factors, which in turn trigger a regenerative response. Here, we review redox paracrine signalling mechanisms in postmitotic tissue regeneration and discuss current challenges and future directions.
- Published
- 2018
439. Mechanisms of Regenerative Potential Activation in Cardiac Mesenchymal Cells.
- Author
-
Docshin PM, Karpov AA, Mametov MV, Ivkin DY, Kostareva AA, and Malashicheva AB
- Abstract
Recovery of the contractile function of the heart and the regeneration of the myocardium after ischemic injury are contemporary issues in regenerative medicine and cell biology. This study aimed to analyze early transcriptional events in cardiac tissue after infarction and to explore the cell population that can be isolated from myocardial tissue. We induced myocardial infarction in Wistar rats by permanent ligation of the left coronary artery and showed a change in the expression pattern of Notch-associated genes and Bmp2/Runx2 in post-MI tissues using RNA sequencing and RT-PCR. We obtained primary cardiac mesenchymal cell (CMC) cultures from postinfarction myocardium by enzymatic dissociation of tissues, which retained part of the activation stimulus and had a pronounced proliferative potential, assessed using a "xCELLigence" real-time system. Hypoxia in vitro also causes healthy CMCs to overexpress Notch-associated genes and Bmp2/Runx2 . Exogenous activation of the Notch signaling pathway by lentiviral transduction of healthy CMCs resulted in a dose-dependent activation of the Runx2 transcription factor but did not affect the activity of the Bmp2 factor. Thus, the results of this study showed that acute hypoxic stress could cause short-term activation of the embryonic signaling pathways Notch and Bmp in CMCs, and this interaction is closely related to the processes of early myocardial remodeling after a heart attack. The ability to correctly modulate and control the corresponding signals in the heart can help increase the regenerative capacity of the myocardium before the formation of fibrotic conditions.
- Published
- 2022
- Full Text
- View/download PDF
440. Cardiac Cell Therapy with Pluripotent Stem Cell-Derived Cardiomyocytes: What Has Been Done and What Remains to Do?
- Author
-
Selvakumar D, Reyes L, and Chong JJH
- Subjects
- Cell Differentiation, Cell- and Tissue-Based Therapy, Humans, Myocardium, Myocytes, Cardiac, Induced Pluripotent Stem Cells, Pluripotent Stem Cells
- Abstract
Purpose of Review: Exciting pre-clinical data presents pluripotent stem cell-derived cardiomyocytes (PSC-CM) as a novel therapeutic prospect following myocardial infarction, and worldwide clinical trials are imminent. However, despite notable advances, several challenges remain. Here, we review PSC-CM pre-clinical studies, identifying key translational hurdles. We further discuss cell production and characterization strategies, identifying markers that may help generate cells which overcome these barriers., Recent Findings: PSC-CMs can robustly repopulate infarcted myocardium with functional, force generating cardiomyocytes. However, current differentiation protocols produce immature and heterogenous cardiomyocytes, creating related issues such as arrhythmogenicity, immunogenicity and poor engraftment. Recent efforts have enhanced our understanding of cardiovascular developmental biology. This knowledge may help implement novel differentiation or gene editing strategies that could overcome these limitations. PSC-CMs are an exciting therapeutic prospect. Despite substantial recent advances, limitations of the technology remain. However, with our continued and increasing biological understanding, these issues are addressable, with several worldwide clinical trials anticipated in the coming years., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
441. MNK2-eIF4E axis promotes cardiac repair in the infarcted mouse heart by activating cyclin D1.
- Author
-
Chen BR, Wei TW, Tang CP, Sun JT, Shan TK, Fan Y, Yang TT, Li YF, Ma Y, Wang SB, Wang ZM, Wang H, Shi JZ, Liu L, Chen JW, Zhou LH, Du C, Sun R, Wang QM, and Wang LS
- Subjects
- Animals, Cyclin D1 metabolism, Mammals metabolism, Mice, Myocytes, Cardiac metabolism, Phosphorylation, Eukaryotic Initiation Factor-4E metabolism, Myocardial Infarction metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
Adult mammals have limited potential for cardiac regeneration after injury. In contrast, neonatal mouse heart, up to 7 days post birth, can completely regenerate after injury. Therefore, identifying the key factors promoting the proliferation of endogenous cardiomyocytes (CMs) is a critical step in the development of cardiac regeneration therapies. In our previous study, we predicted that mitogen-activated protein kinase (MAPK) interacting serine/threonine-protein kinase 2 (MNK2) has the potential of promoting regeneration by using phosphoproteomics and iGPS algorithm. Here, we aimed to clarify the role of MNK2 in cardiac regeneration and explore the underlying mechanism. In vitro, MNK2 overexpression promoted, and MNK2 knockdown suppressed cardiomyocyte proliferation. In vivo, inhibition of MNK2 in CMs impaired myocardial regeneration in neonatal mice. In adult myocardial infarcted mice, MNK2 overexpression in CMs in the infarct border zone activated cardiomyocyte proliferation and improved cardiac repair. In CMs, MNK2 binded to eIF4E and regulated its phosphorylation level. Knockdown of eukaryotic translation initiation factor (eIF4E) impaired the proliferation-promoting effect of MNK2 in CMs. MNK2-eIF4E axis stimulated CMs proliferation by activating cyclin D1. Our study demonstrated that MNK2 kinase played a critical role in cardiac regeneration. Over-expression of MNK2 promoted cardiomyocyte proliferation in vitro and in vivo, at least partly, by activating the eIF4E-cyclin D1 axis. This investigation identified a novel target for heart regenerative therapy., (Copyright © 2022. Published by Elsevier Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
442. Regenerating Endothelium and Restoring Microvascular Endothelial Function.
- Author
-
Hare JM and Yang P
- Subjects
- Endothelium, Humans, Predictive Value of Tests, Endothelium, Vascular, Regeneration
- Abstract
Competing Interests: Funding Support and Author Disclosures Dr Hare has a patent for cardiac cell-based therapy and holds equity in Vestion Inc; maintains a professional relationship with Vestion Inc as a consultant and member of the Board of Directors and Scientific Advisory Board (Vestion Inc did not play a role in the design, conduct, or funding of the study); is Chief Scientific Officer, a compensated consultant, and board member as well as holding equity in Longeveron Inc; and is the co-inventor of intellectual property licensed to Longeveron. Longeveron did not play a role in the design, conduct, or funding of the study. The University of Miami is an equity owner in Longeveron Inc, which has licensed intellectual property from the University of Miami. Dr Yang is a consultant for Terumo Inc; and member of the scientific advisory board for Metcela Inc.
- Published
- 2022
- Full Text
- View/download PDF
443. A small-molecule cocktail promotes mammalian cardiomyocyte proliferation and heart regeneration.
- Author
-
Du J, Zheng L, Gao P, Yang H, Yang WJ, Guo F, Liang R, Feng M, Wang Z, Zhang Z, Bai L, Bu Y, Xing S, Zheng W, Wang X, Quan L, Hu X, Wu H, Chen Z, Chen L, Wei K, Zhang Z, Zhu X, Zhang X, Tu Q, Zhao SM, Lei X, and Xiong JW
- Subjects
- Animals, Cell Proliferation, Heart, Mammals, Rats, Signal Transduction, Zebrafish, Myocardial Infarction drug therapy, Myocytes, Cardiac metabolism
- Abstract
Zebrafish and mammalian neonates possess robust cardiac regeneration via the induction of endogenous cardiomyocyte (CM) proliferation, but adult mammalian hearts have very limited regenerative potential. Developing small molecules for inducing adult mammalian heart regeneration has had limited success. We report a chemical cocktail of five small molecules (5SM) that promote adult CM proliferation and heart regeneration. A high-content chemical screen, along with an algorithm-aided prediction of small-molecule interactions, identified 5SM that efficiently induced CM cell cycle re-entry and cytokinesis. Intraperitoneal delivery of 5SM reversed the loss of heart function, induced CM proliferation, and decreased cardiac fibrosis after rat myocardial infarction. Mechanistically, 5SM potentially targets α1 adrenergic receptor, JAK1, DYRKs, PTEN, and MCT1 and is connected to lactate-LacRS2 signaling, leading to CM metabolic switching toward glycolysis/biosynthesis and CM de-differentiation before entering the cell-cycle. Our work sheds lights on the understanding CM regenerative mechanisms and opens therapeutic avenues for repairing the heart., Competing Interests: Declaration of interests The authors declare no competing financial interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
444. Bacterial Cellulose and ECM Hydrogels: An Innovative Approach for Cardiovascular Regenerative Medicine.
- Author
-
Silva IGRD, Pantoja BTDS, Almeida GHDR, Carreira ACO, and Miglino MA
- Subjects
- Animals, Cats, Cellulose metabolism, Dogs, Extracellular Matrix metabolism, Hydrogels chemistry, Hydrogels therapeutic use, Quality of Life, Regenerative Medicine, Tissue Engineering, Cat Diseases, Dog Diseases metabolism
- Abstract
Cardiovascular diseases are considered the leading cause of death in the world, accounting for approximately 85% of sudden death cases. In dogs and cats, sudden cardiac death occurs commonly, despite the scarcity of available pathophysiological and prevalence data. Conventional treatments are not able to treat injured myocardium. Despite advances in cardiac therapy in recent decades, transplantation remains the gold standard treatment for most heart diseases in humans. In veterinary medicine, therapy seeks to control clinical signs, delay the evolution of the disease and provide a better quality of life, although transplantation is the ideal treatment. Both human and veterinary medicine face major challenges regarding the transplantation process, although each area presents different realities. In this context, it is necessary to search for alternative methods that overcome the recovery deficiency of injured myocardial tissue. Application of biomaterials is one of the most innovative treatments for heart regeneration, involving the use of hydrogels from decellularized extracellular matrix, and their association with nanomaterials, such as alginate, chitosan, hyaluronic acid and gelatin. A promising material is bacterial cellulose hydrogel, due to its nanostructure and morphology being similar to collagen. Cellulose provides support and immobilization of cells, which can result in better cell adhesion, growth and proliferation, making it a safe and innovative material for cardiovascular repair.
- Published
- 2022
- Full Text
- View/download PDF
445. Measuring cardiomyocyte cell-cycle activity and proliferation in the age of heart regeneration.
- Author
-
Auchampach J, Han L, Huang GN, Kühn B, Lough JW, O'Meara CC, Payumo AY, Rosenthal NA, Sucov HM, Yutzey KE, and Patterson M
- Subjects
- Animals, Cell Cycle, Cell Proliferation, Heart physiology, Mammals, Myocytes, Cardiac physiology, Regeneration
- Abstract
During the past two decades, the field of mammalian myocardial regeneration has grown dramatically, and with this expanded interest comes increasing claims of experimental manipulations that mediate bona fide proliferation of cardiomyocytes. Too often, however, insufficient evidence or improper controls are provided to support claims that cardiomyocytes have definitively proliferated, a process that should be strictly defined as the generation of two de novo functional cardiomyocytes from one original cardiomyocyte. Throughout the literature, one finds inconsistent levels of experimental rigor applied, and frequently the specific data supplied as evidence of cardiomyocyte proliferation simply indicate cell-cycle activation or DNA synthesis, which do not necessarily lead to the generation of new cardiomyocytes. In this review, we highlight potential problems and limitations faced when characterizing cardiomyocyte proliferation in the mammalian heart, and summarize tools and experimental standards, which should be used to support claims of proliferation-based remuscularization. In the end, definitive establishment of de novo cardiomyogenesis can be difficult to prove; therefore, rigorous experimental strategies should be used for such claims.
- Published
- 2022
- Full Text
- View/download PDF
446. Injectable Extracellular Matrix Microparticles Promote Heart Regeneration in Mice with Post-ischemic Heart Injury.
- Author
-
Wang X, Ansari A, Pierre V, Young K, Kothapalli CR, von Recum HA, and Senyo SE
- Subjects
- Animals, Biocompatible Materials metabolism, Biocompatible Materials pharmacology, Hydrogels metabolism, Mice, Regeneration, Swine, Tissue Engineering methods, Extracellular Matrix metabolism, Heart Injuries metabolism
- Abstract
Ischemic heart injury causes permanent cardiomyocyte loss and fibrosis impairing cardiac function. Tissue derived biomaterials have shown promise as an injectable treatment for the post-ischemic heart. Specifically, decellularized extracellular matrix (dECM) is a protein rich suspension that forms a therapeutic hydrogel once injected and improves the heart post-injury response in rodents and pig models. Current dECM-derived biomaterials are delivered to the heart as a liquid dECM hydrogel precursor or colloidal suspension, which gels over several minutes. To increase the functionality of the dECM therapy, an injectable solid dECM microparticle formulation derived from heart tissue to control sizing and extend stability in aqueous conditions is developed. When delivered into the infarcted mouse heart, these dECM microparticles protect cardiac function promote vessel density and reduce left ventricular remodeling by sustained delivery of biomolecules. Longer retention, higher stiffness, and slower protein release of dECM microparticles are noted compared to liquid dECM hydrogel precursor. In addition, the dECM microparticle can be developed as a platform for macromolecule delivery. Together, the results suggest that dECM microparticles can be developed as a modular therapy for heart injury., (© 2022 Wiley-VCH GmbH.)
- Published
- 2022
- Full Text
- View/download PDF
447. Induction of Stem-Cell-Derived Cardiomyogenesis by Fibroblast Growth Factor 10 (FGF10) and Its Interplay with Cardiotrophin-1 (CT-1).
- Author
-
Khosravi F, Ahmadvand N, Wartenberg M, and Sauer H
- Abstract
For heart regeneration purposes, embryonic stem cell (ES)-based strategies have been developed to induce the proliferation of cardiac progenitor cells towards cardiomyocytes. Fibroblast growth factor 10 (FGF10) contributes to cardiac development and induces cardiomyocyte differentiation in vitro. Yet, among pro-cardiogenic factors, including cardiotrophin-1 (CT-1), the hyperplastic function of FGF10 in cardiomyocyte turnover remains to be further characterized. We investigated the proliferative effects of FGF10 on ES-derived cardiac progenitor cells in the intermediate developmental stage and examined the putative interplay between FGF10 and CT-1 in cardiomyocyte proliferation. Mouse ES cells were treated with FGF10 and/or CT-1. Differential expression of cardiomyocyte-specific gene markers was analyzed at transcript and protein levels. Substantial upregulation of sarcomeric α-actinin was detected by qPCR, flow cytometry, Western blot and immunocytochemistry. FGF10 enhanced the expression of other structural proteins (MLC-2a, MLC-2v and TNNT2), transcriptional factors (NKX2-5 and GATA4), and proliferation markers (Aurora B and YAP-1). FGF10/CT-1 co-administration led to an upregulation of proliferation markers, suggesting the synergistic potential of FGF10 + CT-1 on cardiomyogenesis. In summary, we provided evidence that FGF10 and CT-1 induce cardiomyocyte structural proteins, associated transcription factors, and cardiac cell proliferation, which could be applicable in therapies to replenish damaged cardiomyocytes.
- Published
- 2022
- Full Text
- View/download PDF
448. Cardiac regeneration following myocardial infarction: the need for regeneration and a review of cardiac stromal cell populations used for transplantation.
- Author
-
Alonaizan R and Carr C
- Subjects
- Humans, Myocytes, Cardiac metabolism, Regeneration, Stromal Cells metabolism, Myocardial Infarction metabolism, Myocardial Infarction therapy, Stem Cell Transplantation
- Abstract
Myocardial infarction is a leading cause of death globally due to the inability of the adult human heart to regenerate after injury. Cell therapy using cardiac-derived progenitor populations emerged about two decades ago with the aim of replacing cells lost after ischaemic injury. Despite early promise from rodent studies, administration of these populations has not translated to the clinic. We will discuss the need for cardiac regeneration and review the debate surrounding how cardiac progenitor populations exert a therapeutic effect following transplantation into the heart, including their ability to form de novo cardiomyocytes and the release of paracrine factors. We will also discuss limitations hindering the cell therapy field, which include the challenges of performing cell-based clinical trials and the low retention of administered cells, and how future research may overcome them., (© 2022 The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
449. Heart regeneration: 20 years of progress and renewed optimism.
- Author
-
Garbern JC and Lee RT
- Subjects
- Animals, Humans, Stem Cell Transplantation methods, Heart physiology, Myocardium cytology, Myocytes, Cardiac cytology, Regeneration physiology, Regenerative Medicine methods
- Abstract
Cardiovascular disease is a leading cause of death worldwide, and thus there remains great interest in regenerative approaches to treat heart failure. In the past 20 years, the field of heart regeneration has entered a renaissance period with remarkable progress in the understanding of endogenous heart regeneration, stem cell differentiation for exogenous cell therapy, and cell-delivery methods. In this review, we highlight how this new understanding can lead to viable strategies for human therapy. For the near term, drugs, electrical and mechanical devices, and heart transplantation will remain mainstays of cardiac therapies, but eventually regenerative therapies based on fundamental regenerative biology may offer more permanent solutions for patients with heart failure., Competing Interests: Declaration of interests R.T.L. is a co-founder, scientific advisory board member, and private equity holder of Elevian, R.T.L. is a member of the scientific advisory board of Revidia Therapeutics and a consultant to BlueRock Therapeutics., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
450. Identification of enhancer regulatory elements that direct epicardial gene expression during zebrafish heart regeneration.
- Author
-
Cao Y, Xia Y, Balowski JJ, Ou J, Song L, Safi A, Curtis T, Crawford GE, Poss KD, and Cao J
- Subjects
- Animals, Animals, Genetically Modified genetics, Animals, Genetically Modified metabolism, Chromatin metabolism, GTP-Binding Protein alpha Subunits, Gi-Go genetics, GTP-Binding Protein alpha Subunits, Gi-Go metabolism, Gene Expression Regulation, Larva growth & development, Larva metabolism, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Neural Cell Adhesion Molecules genetics, Neural Cell Adhesion Molecules metabolism, Pericardium cytology, Zebrafish growth & development, Zebrafish metabolism, Zebrafish Proteins genetics, Zebrafish Proteins metabolism, Enhancer Elements, Genetic genetics, Heart physiology, Pericardium metabolism, Regeneration physiology
- Abstract
The epicardium is a mesothelial tissue layer that envelops the heart. Cardiac injury activates dynamic gene expression programs in epicardial tissue, which in zebrafish enables subsequent regeneration through paracrine and vascularizing effects. To identify tissue regeneration enhancer elements (TREEs) that control injury-induced epicardial gene expression during heart regeneration, we profiled transcriptomes and chromatin accessibility in epicardial cells purified from regenerating zebrafish hearts. We identified hundreds of candidate TREEs, which are defined by increased chromatin accessibility of non-coding elements near genes with increased expression during regeneration. Several of these candidate TREEs were incorporated into stable transgenic lines, with five out of six elements directing injury-induced epicardial expression but not ontogenetic epicardial expression in larval hearts. Whereas two independent TREEs linked to the gene gnai3 showed similar functional features of gene regulation in transgenic lines, two independent ncam1a-linked TREEs directed distinct spatiotemporal domains of epicardial gene expression. Thus, multiple TREEs linked to a regeneration gene can possess either matching or complementary regulatory controls. Our study provides a new resource and principles for understanding the regulation of epicardial genetic programs during heart regeneration. This article has an associated 'The people behind the papers' interview., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2022. Published by The Company of Biologists Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.