Pubblicazioni recenti - cardiac stem
-
Melatonin and prolonged physical activity attenuated the detrimental effects of diabetic condition on murine cardiac tissue.
Tissue Cell2021 Jan;69():101486. doi: S0040-8166(21)00002-1.
Rahbarghazi Afshin, Siahkouhian Marefat, Rahbarghazi Reza, Ahmadi Mahdi, Bolboli Lotfali, Mahdipour Mahdi, Haghighi Leila, Hassanpour Mehdi, Sokouti Nasimi Fatemeh, Keyhanmanesh Rana,
Abstract
In this study, the combined effects of four-week swimming training and melatonin were examined on the oxidative response, inflammation, apoptosis, and angiogenesis capacity of cardiac tissue in the mouse model of diabetes. The mice were randomly allocated into five groups (n = 10 per group) as follows: Control; Diabetic group; Diabetic + Melatonin group; Diabetic + Exercise group; and Diabetic + Exercise + Melatonin group. 50 mg/kg streptozotocin was intraperitoneally administrated. In melatonin-treated groups, melatonin was injected intraperitoneally at 3 mg/kg body weight for four weeks and twice weekly. Swimming exercises were performed for four weeks. We measured cardiac superoxide dismutase, glutathione peroxidase enzymes, malondialdehyde, and total antioxidant capacity. The expression of tumor necrosis factor-?, Caspase?3, Sirtuin1, and Connexin-43 was measured using real-time PCR analysis. The vascular density was analyzed by immunohistochemistry using CD31 and ?-smooth muscle actin antibodies. The combination of melatonin and exercise elevated cardiac superoxide dismutase, glutathione peroxidase coincided with the reduction of malondialdehyde and increase of total antioxidant capacity as compared to the diabetic mice (p < 0.05). In Diabetic + Exercise + Melatonin mice, tumor necrosis factor-?, Caspase?3 was significantly down-regulated compared to the Diabetic group (p < 0.05). Melatonin and exercise suppressed the expression of Connexin-43 and Sirtuin1 in diabetic mice in comparison with the control mice (p < 0.05). H & E staining showed necrosis and focal hyperemia reduction in the Diabetic + Exercise + Melatonin group compared to the Diabetic group. Data showed a decrease of CD31 and ?-smooth muscle actin vessels in the Diabetic group as compared to the normal samples (p < 0.05). The number of CD31 vessels, but not ?-smooth muscle actin type, increased in the Diabetic + Exercise + Melatonin group compared to the Diabetic mice. These data demonstrated that exercise along with melatonin administration could diminish the detrimental effects of diabetes on cardiac tissue via using different mechanisms.
Copyright © 2021 Elsevier Ltd. All rights reserved.
Guarda su PubMed -
Cultured Cardiac Fibroblasts and Myofibroblasts Express Sushi Containing Domain 2 and Assemble a Unique Fibronectin Rich Matrix.
Exp Cell Res2021 Jan;():112489. doi: S0014-4827(21)00020-3.
Schmuck Eric G, Roy Sushmita, Dhillon Anisa, Walker Sydney, Spinali Keith, Colevas Sophia, Zhou Tianhua, Chhabra Gagan, Liu Yuming, Kader Sagar Md Abdul, Childs Charlie J, Kink John A, Eliceiri Kevin W, Hematti Peiman, Raval Amish N,
Abstract
Cardiac fibroblasts and myofibroblasts assemble and maintain extracellular matrix during normal development and following injury. Culture expansion of these cells yield a bioengineered matrix that could lead to intriguing therapeutic opportunities. For example, we reported that cultured rat cardiac fibroblasts form a matrix that can be used to delivery therapeutic stem cells. Furthermore, we reported that matrix derived from cultured human cardiac fibroblasts/myofibroblasts converted monocytes into macrophages that express interesting anti-inflammatory and pro-angiogenic properties. Expanding these matrix investigations require characterization of the source cells for quality control. In these efforts, we observed and herein report that Sushi Containing Domain 2 (SUSD2) is a novel and consistent marker for cultured human cardiac fibroblast and myofibroblasts.
Copyright © 2021. Published by Elsevier Inc.
Guarda su PubMed -
Knockout of MYOM1 in human cardiomyocytes leads to myocardial atrophy via impairing calcium homeostasis.
J Cell Mol Med2021 Jan;():. doi: 10.1111/jcmm.16268.
Hang Chengwen, Song Yuanxiu, Li Ya'nan, Zhang Siyao, Chang Yun, Bai Rui, Saleem Amina, Jiang Mengqi, Lu Wenjing, Lan Feng, Cui Ming,
Abstract
Myomesin-1 (encoded by MYOM1 gene) is expressed in almost all cross-striated muscles, whose family (together with myomesin-2 and myomesin-3) helps to cross-link adjacent myosin to form the M-line in myofibrils. However, little is known about its biological function, causal relationship and mechanisms underlying the MYOM1-related myopathies (especially in the heart). Regrettably, there is no MYMO1 knockout model for its study so far. A better and further understanding of MYOM1 biology is urgently needed. Here, we used CRISPR/Cas9 gene-editing technology to establish an MYOM1 knockout human embryonic stem cell line (MYOM1 hESC), which was then differentiated into myomesin-1 deficient cardiomyocytes (MYOM1 hESC-CMs) in vitro. We found that myomesin-1 plays an important role in sarcomere assembly, contractility regulation and cardiomyocytes development. Moreover, myomesin-1-deficient hESC-CMs can recapitulate myocardial atrophy phenotype in vitro. Based on this model, not only the biological function of MYOM1, but also the aetiology, pathogenesis, and potential treatments of myocardial atrophy caused by myomesin-1 deficiency can be studied.
© 2021 The Authors. Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd.
Guarda su PubMed -
Myocyte-specific enhancer factor 2c triggers transdifferentiation of adipose tissue-derived stromal cells into spontaneously beating cardiomyocyte-like cells.
Sci Rep2021 Jan;11(1):1520. doi: 10.1038/s41598-020-80848-3.
Takashima Shinichiro, Usui Soichiro, Inoue Oto, Goten Chiaki, Yamaguchi Kosei, Takeda Yusuke, Cui Shihe, Sakai Yoshio, Hayashi Kenshi, Sakata Kenji, Kawashiri Masa-Aki, Takamura Masayuki,
Abstract
Cardiomyocyte regeneration is limited in adults. The adipose tissue-derived stromal vascular fraction (Ad-SVF) contains pluripotent stem cells that rarely transdifferentiate into spontaneously beating cardiomyocyte-like cells (beating CMs). However, the characteristics of beating CMs and the factors that regulate the differentiation of Ad-SVF toward the cardiac lineage are unknown. We developed a simple culture protocol under which the adult murine inguinal Ad-SVF reproducibly transdifferentiates into beating CMs without induction. The beating CMs showed the striated ventricular phenotype of cardiomyocytes and synchronised oscillation of the intracellular calcium concentration among cells on day 28 of Ad-SVF primary culture. We also identified beating CM-fated progenitors (CFPs) and performed single-cell transcriptome analysis of these CFPs. Among 491 transcription factors that were differentially expressed (??1.75-fold) in CFPs and the beating CMs, myocyte-specific enhancer 2c (Mef2c) was key. Transduction of Ad-SVF cells with Mef2c using a lentiviral vector yielded CFPs and beating CMs with?~?tenfold higher cardiac troponin T expression, which was abolished by silencing of Mef2c. Thus, we identified the master gene required for transdifferentiation of Ad-SVF into beating CMs. These findings will facilitate the development of novel cardiac regeneration therapies based on gene-modified, cardiac lineage-directed Ad-SVF cells.
Guarda su PubMed -
Proteomics of Asrij Perturbation in Drosophila Lymph Glands for Identification of New Regulators of Hematopoiesis.
Mol Cell Proteomics2019 Jun;18(6):1171-1182. doi: S1535-9476(20)31818-1.
Sinha Saloni, Ray Arindam, Abhilash Lakshman, Kumar Manish, Sreenivasamurthy Sreelakshmi K, Keshava Prasad T S, Inamdar Maneesha S,
Abstract
Hematopoiesis is the process of differentiation of precursor blood cells into mature blood cells that is controlled by a complex set of molecular interactions. Understanding hematopoiesis is important for the study of hematological disorders. However, a comprehensive understanding of how physiological and genetic mechanisms regulate blood cell precursor maintenance and differentiation is lacking. Owing to simplicity and ease of genetic analysis, the Drosophila melanogaster lymph gland (LG) is an excellent model to study hematopoiesis. Here, we quantitatively analyzed the LG proteome under genetic conditions that either maintain precursors or promote their differentiation in vivo, by perturbing expression of Asrij, a conserved endosomal regulator of hematopoiesis. Using iTRAQ-based quantitative proteomics, we determined the relative expression levels of proteins in Asrij-knockout and overexpressing LGs from 1500 larval dissections compared with wild type. Our data showed that at least 6.5% of the Drosophila proteome is expressed in wild type LGs. Of the 2133 proteins identified, 780 and 208 proteins were common to previously reported cardiac tube and hemolymph proteomes, respectively, resulting in the identification of 1238 proteins exclusive to the LG. Perturbation of Asrij levels led to differential expression of 619 proteins, of which 27% have human homologs implicated in various diseases. Proteins regulating metabolism, immune system, signal transduction and vesicle-mediated transport were significantly enriched. Immunostaining of representative candidates from the enriched categories and previous reports confirmed 73% of our results, indicating the validity of our LG proteome. Our study provides, for the first time, an in vivo proteomics resource for identifying novel regulators of hematopoiesis that will also be applicable to understanding vertebrate blood cell development.
Copyright © 2019 © 2019 Sinha et al. Published by Elsevier Inc. All rights reserved.
Guarda su PubMed -
The influence of melatonin on the heart rhythm - An in vitro simulation with murine embryonic stem cell derived cardiomyocytes.
Biomed Pharmacother2021 Jan;136():111245. doi: S0753-3322(21)00030-5.
Niehoff Julius, Matzkies Matthias, Nguemo Filomain, Hescheler Jürgen, Reppel Michael,
Abstract
BACKGROUND:
In healthy individuals, a major factor influencing the heart rate variability (HRV) is the circadian rhythm. The role of melatonin as an essential component of the circadian rhythm in the adult human organism and the beneficial effects of a treatment with melatonin during the fetal period is well described. Toxic effects of melatonin are discussed less frequently. Since pharmacological studies cannot be carried out on pregnant women, the establishment of an equivalent in vitro model is important. We therefore tested whether melatonin can influence the beat rate variability (BRV) of spontaneously beating cardiomyocytes derived from murine embryonic stem cells (mESCs) and whether melatonin exhibits toxic effects in this in vitro model.
METHODS:
Microelectrode Arrays recorded extracellular field potentials of spontaneously beating cardiomyocytes. Melatonin was applied in a concentration range from 10 M to 10 M. The analysis of the BRV focused on time domain methods.
RESULTS:
In line with clinical observations, melatonin decreased the beating frequency and increased the BRV. The effect of melatonin up to a concentration of 10 M was reversible, whereas the application of higher concentrations induced an irreversible effect.
CONCLUSION:
The study underlines the potential of this in vitro model to help explore the development of circadian rhythms and their modulation by melatonin in the embryonic phase. The results imply that melatonin influences the heart rhythm as early as during the embryonic heart development. Furthermore, the results indicate a potentially toxic effect of melatonin that has not been described in detail before.
Copyright © 2021. Published by Elsevier Masson SAS.
Guarda su PubMed -
Embryo-toxicity of docosahexaenoic and eicosapentaenoic acids: In vivo and in silico investigations using the chick embryo model.
Biomed Pharmacother2021 Jan;136():111218. doi: S0753-3322(21)00003-2.
Salari Zohreh, Tavakkoli Hadi, Khosravi Ahmad, Karamad Elahe, Salarkia Ehsan, Ansari Mehdi, Dabiri Shahriar, Mortazaeizdeh Abbas, Mosallanejad Seyedeh Saedeh, Sharifi Fatemeh,
Abstract
OBJECTIVE:
The objective of the current study was to evaluate the embryo-toxicity of omega-3 fatty acids.
METHODS:
Firstly, the embryo-toxicity of docosahexaenoic (DHA) and eicosapentaenoic acids (EPA), as well as their interaction with Bcl-2 family members, were predicted using an in silico assay. In the next step, the embryonic pathological lesions and amniotic fluid biochemical changes following omega-3 treatment were investigated using a chick embryo model. Finally, the drug's vascular apoptotic effect on the chick's yolk sac membrane (YSM) was assessed.
RESULTS:
In silico simulations revealed the embryo-toxicity, tissue-toxicity (respiratory and cardiovascular), and vascular-toxicity (apoptotic activity) of DHA and EPA. There was also an accurate interaction between DHA and EPA with Bax (Binding affinity: -7.6 and -10.6 kcal/mol) and Bcl-2 (Binding affinity: -8.0 and -12.2 kcal/mol), respectively. Moreover, DHA and EPA administrations were related to various adverse consequences, including weight loss and lesions in the respiratory and cardiovascular systems. Histopathological findings consisted of pulmonary edema, airway dilatation, increased interstitial tissue, and hyperemia in the lungs, heart, liver, kidney, and brain. Morphometric evaluation of the YSM vasculature revealed that the vascular apoptotic effect of omega-3was associated with a significant reduction in mean capillary area. In immunohistochemistry assay, increased expression of BAX and low expression of Bcl-2 affirmed apoptosis in YSM vessels.
CONCLUSION:
According to the results of this study, one could confirm that the possible embryo-toxicity of omega-3 was approved by data presented in this research. The obtained results also support the suspicion that alteration of the apoptotic-related proteins in vessels is an essential pathway in embryo-toxicity of omega-3.
Copyright © 2021 The Author(s). Published by Elsevier Masson SAS.. All rights reserved.
Guarda su PubMed -
Relationships Between Constituents of Energy Drinks and Beating Parameters in Human Induced Pluripotent Cell (iPSC)-Derived Cardiomyocytes.
Food Chem Toxicol2021 Jan;():111979. doi: S0278-6915(21)00013-2.
Luo Yu-Syuan, Chen Zunwei, Blanchette Alexander D, Zhou Yi-Hui, Wright Fred A, Baker Erin S, Chiu Weihsueh A, Rusyn Ivan,
Abstract
Consumption of energy drinks has been associated with adverse cardiovascular effects; however, little is known about the ingredients that may contribute to these effects. We therefore characterized the chemical profiles and in vitro effects of energy drinks and their ingredients on human induced pluripotent stem cell (iPSC)-derived cardiomyocytes, and identified the putative active ingredients using a multivariate prediction model. Energy drinks from 17 widely-available over-the-counter brands were evaluated in this study. The concentrations of six common molecular ingredients (caffeine, taurine, riboflavin, pantothenic acid, adenine, and L-methionine) were quantified by coupling liquid chromatography with a triple quadrupole mass spectrometer for the acquisition of LC-MS/MS spectra. In addition, untargeted analyses for each beverage were performed with a platform combining LC, ion mobility spectrometry and mass spectrometry (LC-IMS-MS) measurements. Approximately 300 features were observed per sample in the untargeted studies, and of these ?100 were identified. In vitro effects of energy drinks and some of their molecular ingredients were then tested in iPSC-derived cardiomyocytes. Data on the beat rate (positive and negative chronotropy), ion channel function (QT prolongation), and cytotoxicity were collected in a dilution series. We found that some of the energy drinks elicited adverse effects on the cardiomyocytes with the most common being an increase in the beat rate, while QT prolongation was also observed at the lowest concentrations. Finally, concentration addition modeling using quantitative data from the 6 common ingredients and multivariate prediction modeling was used to determine potential molecular ingredients responsible for the adverse effects on the cardiomyocytes. These analyses suggested theophylline, adenine, and azelate as possibly contributing to the in vitro effects of energy drinks on QT prolongation in cardiomyocytes.
Copyright © 2021 Elsevier Ltd. All rights reserved.
Guarda su PubMed -
To be or not to be: endothelial cell plasticity in development, repair, and disease.
Angiogenesis2021 Jan;():. doi: 10.1007/s10456-020-09761-7.
Greenspan Leah J, Weinstein Brant M,
Abstract
Endothelial cells display an extraordinary plasticity both during development and throughout adult life. During early development, endothelial cells assume arterial, venous, or lymphatic identity, while selected endothelial cells undergo additional fate changes to become hematopoietic progenitor, cardiac valve, and other cell types. Adult endothelial cells are some of the longest-lived cells in the body and their participation as stable components of the vascular wall is critical for the proper function of both the circulatory and lymphatic systems, yet these cells also display a remarkable capacity to undergo changes in their differentiated identity during injury, disease, and even normal physiological changes in the vasculature. Here, we discuss how endothelial cells become specified during development as arterial, venous, or lymphatic endothelial cells or convert into hematopoietic stem and progenitor cells or cardiac valve cells. We compare findings from in vitro and in vivo studies with a focus on the zebrafish as a valuable model for exploring the signaling pathways and environmental cues that drive these transitions. We also discuss how endothelial plasticity can aid in revascularization and repair of tissue after damage- but may have detrimental consequences under disease conditions. By better understanding endothelial plasticity and the mechanisms underlying endothelial fate transitions, we can begin to explore new therapeutic avenues.
Guarda su PubMed -
Use of hiPSC to explicate genomic predisposition to anthracycline-induced cardiotoxicity.
Pharmacogenomics2021 Jan;():. doi: 10.2217/pgs-2020-0104.
Magdy Tarek, Burridge Paul W,
Abstract
The anticancer agents of the anthracycline family are commonly associated with the potential to cause severe toxicity to the heart. To solve the question of why particular a patient is predisposed to anthracycline-induced cardiotoxicity (AIC), researchers have conducted numerous pharmacogenomic studies and identified more than 60 loci associated with AIC. To date, none of these identified loci have been developed into US FDA-approved biomarkers for use in routine clinical practice. With advances in the application of human-induced pluripotent stem cell-derived cardiomyocytes, sequencing technologies and genomic editing techniques, variants associated with AIC can now be validated in a human model. Here, we provide a comprehensive overview of known genetic variants associated with AIC from the perspective of how human-induced pluripotent stem cell-derived cardiomyocytes can be used to help better explain the genomic predilection to AIC.
Guarda su PubMed -
Zebra-Fishing for Regenerative Awakening in Mammals.
Biomedicines2021 Jan;9(1):. doi: E65.
Massoz Laura, Dupont Marie Alice, Manfroid Isabelle,
Abstract
Regeneration is defined as the ability to regrow an organ or a tissue destroyed by degeneration or injury. Many human degenerative diseases and pathologies, currently incurable, could be cured if functional tissues or cells could be restored. Unfortunately, humans and more generally mammals have limited regenerative capabilities, capacities that are even further declining with age, contrary to simpler organisms. Initially thought to be lost during evolution, several studies have revealed that regenerative mechanisms are still present in mammals but are latent and thus they could be stimulated. To do so there is a pressing need to identify the fundamental mechanisms of regeneration in species able to efficiently regenerate. Thanks to its ability to regenerate most of its organs and tissues, the zebrafish has become a powerful model organism in regenerative biology and has recently engendered a number of studies attesting the validity of awakening the regenerative potential in mammals. In this review we highlight studies, particularly in the liver, pancreas, retina, heart, brain and spinal cord, which have identified conserved regenerative molecular events that proved to be beneficial to restore murine and even human cells and which helped clarify the real clinical translation potential of zebrafish research to mammals.
Guarda su PubMed -
Clinical Considerations for a Family with Dilated Cardiomyopathy, Sudden Cardiac Death, and a Novel Frameshift Mutation.
Int J Mol Sci2021 Jan;22(2):. doi: E670.
Micaglio Emanuele, Monasky Michelle M, Bernardini Andrea, Mecarocci Valerio, Borrelli Valeria, Ciconte Giuseppe, Locati Emanuela T, Piccoli Marco, Ghiroldi Andrea, Anastasia Luigi, Pappone Carlo,
Abstract
Dilated cardiomyopathy (DCM) is the leading indication for heart transplantation. gene truncating mutations account for about 25% of familial DCM cases and for 18% of sporadic DCM cases. The clinical relevance of specific variants in has been difficult to determine because of the sheer size of the protein for which encodes, as well as existing extensive genetic variation. Clinicians should communicate novel clinically-relevant variants and genotype-phenotype associations, so that animal studies evaluating the molecular mechanisms are always conducted with a focus on clinical significance. In the present study, we report for the first time the novel truncating heterozygous variant NM_001256850.1:c.72777_72783del (p.Phe24259Leufs*51) in the gene and its association with DCM in a family with sudden death. This variant occurs in the A-band region of the sarcomere, in a known mutational hotspot of the gene. Truncating titin variants that occur in this region are the most common cause of DCM and have been rarely reported in asymptomatic individuals, differently from other pathogenic gene variants. Further studies are warranted to better understand this particular clinically-relevant variant.
Guarda su PubMed -
The FGF-AKT pathway is necessary for cardiomyocyte survival for heart regeneration in zebrafish.
Dev Biol2021 Jan;():. doi: S0012-1606(21)00003-8.
Tahara Naoyuki, Akiyama Ryutaro, Wang Justin, Kawakami Hiroko, Bessho Yasumasa, Kawakami Yasuhiko,
Abstract
Zebrafish have a remarkable ability to regenerate the myocardium after injury by proliferation of pre-existing cardiomyocytes. Fibroblast growth factor (FGF) signaling is known to play a critical role in zebrafish heart regeneration through promotion of neovascularization of the regenerating myocardium. Here, we define an additional function of FGF signaling in the zebrafish myocardium after injury. We find that FGF signaling is active in a small fraction of cardiomyocytes before injury, and that the number of FGF signaling-positive cardiomyocytes increases after amputation-induced injury. We show that ERK phosphorylation is prominent in endothelial cells, but not in cardiomyocytes. In contrast, basal levels of phospho-AKT positive cardiomyocytes are detected before injury, and the ratio of phosphorylated AKT-positive cardiomyocytes increases after injury, indicating a role of AKT signaling in cardiomyocytes following injury. Inhibition of FGF signaling reduced the number of phosphorylated AKT-positive cardiomyocytes and increased cardiomyocyte death without injury. Heart injury did not induce cardiomyocyte death; however, heart injury in combination with inhibition of FGF signaling caused significant increase in cardiomyocyte death. Pharmacological inhibition of AKT signaling after heart injury also caused increased cardiomyocyte death. Our data support the idea that FGF-AKT signaling-dependent cardiomyocyte survival is necessary for subsequent heart regeneration.
Copyright © 2021. Published by Elsevier Inc.
Guarda su PubMed -
Prediction of putative small molecules for manipulation of enriched signalling pathways in hESC-derived early cardiovascular progenitors by bioinformatics analysis.
IET Syst Biol2019 Apr;13(2):77-83. doi: 10.1049/iet-syb.2018.5037.
Vahdat Sadaf, Bakhshandeh Behnaz,
Abstract
Human pluripotent stem cell-derived cardiovascular progenitor cells (CPCs) are considered as powerful tools for cardiac regenerative medicine and developmental study. Mesoderm posterior1 (MESP1 ) cells are identified as the earliest CPCs from which almost all cardiac cell types are generated. Molecular insights to the transcriptional regulatory factors of early CPCs are required to control cell fate decisions. Herein, the microarray data set of human embryonic stem cells (hESCs)-derived MESP1 cells was analysed and differentially expressed genes (DEGs) were identified in comparison to undifferentiated hESCs and MESP1-negative cells. Then, gene ontology and pathway enrichment analysis of DEGs were carried out with the subsequent prediction of putative regulatory small molecules for modulation of CPC fate. Some key signalling cascades of cardiogenesis including Hippo, Wnt, transforming growth factor-?, and PI3K/Akt were highlighted in MESP1 cells. The transcriptional regulatory network of MESP1 cells were visualised through interaction networks of DEGs. Additionally, 35 promising chemicals were predicted based on correlations with gene expression signature of MESP1 cells for effective in vitro CPC manipulation. Studying the transcriptional profile of MESP1 cells resulted into the identification of important signalling pathways and chemicals which could be introduced as powerful tools to manage proliferation and differentiation of hESC-derived CPCs more efficiently.
© 2019 The Institution of Engineering and Technology.
Guarda su PubMed -
Oxygen Is an Ambivalent Factor for the Differentiation of Human Pluripotent Stem Cells in Cardiac 2D Monolayer and 3D Cardiac Spheroids.
Int J Mol Sci2021 Jan;22(2):. doi: E662.
Souidi Monia, Sleiman Yvonne, Acimovic Ivana, Pribyl Jan, Charrabi Azzouz, Baecker Volker, Scheuermann Valerie, Pesl Martin, Jelinkova Sarka, Skladal Petr, Dvorak Petr, Lacampagne Alain, Rotrekl Vladimir, Meli Albano C,
Abstract
Numerous protocols of cardiac differentiation have been established by essentially focusing on specific growth factors on human pluripotent stem cell (hPSC) differentiation efficiency. However, the optimal environmental factors to obtain cardiac myocytes in network are still unclear. The mesoderm germ layer differentiation is known to be enhanced by low oxygen exposure. Here, we hypothesized that low oxygen exposure enhances the molecular and functional maturity of the cardiomyocytes. We aimed at comparing the molecular and functional consequences of low (5% O or LOE) and high oxygen exposure (21% O or HOE) on cardiac differentiation of hPSCs in 2D- and 3D-based protocols. hPSC-CMs were differentiated through both the 2D (monolayer) and 3D (embryoid body) protocols using several lines. Cardiac marker expression and cell morphology were assessed. The mitochondrial localization and metabolic properties were evaluated. The intracellular Ca handling and contractile properties were also monitored. The 2D cardiac monolayer can only be differentiated in HOE. The 3D cardiac spheroids containing hPSC-CMs in LOE further exhibited cardiac markers, hypertrophy, steadier SR Ca release properties revealing a better SR Ca handling, and enhanced contractile force. Preserved distribution of mitochondria and similar oxygen consumption by the mitochondrial respiratory chain complexes were also observed. Our results brought evidences that LOE is moderately beneficial for the 3D cardiac spheroids with hPSC-CMs exhibiting further maturity. In contrast, the 2D cardiac monolayers strictly require HOE.
Guarda su PubMed -
Fibroblast Growth Factor-1 Released from a Heparin Coacervate Improves Cardiac Function in a Mouse Myocardial Infarction Model.
ACS Biomater Sci Eng2017 Sep;3(9):1988-1999. doi: 10.1021/acsbiomaterials.6b00509.
Wang Zhouguang, Long Daniel W, Huang Yan, Khor Sinan, Li Xiaokun, Jian Xiao, Wang Yadong,
Abstract
Emerging evidence supports the beneficial effect of fibroblast growth factor-1 (FGF1) on heart diseases, but its application has been hindered by the short half-life and limited bioactivity of the free protein. We designed an injectable coacervate to facilitate robust growth factor delivery, which would both protect and increase the bioactivity of growth factors. In this study, a model for acute myocardial infarction was established in mice, and the cardioprotective effect of the FGF1 coacervate was investigated. Echocardiographic results showed that the FGF1 coacervate inhibited ventricular dilation and preserved cardiac contractibility more than the free FGF1 and the saline control within the 6-week duration of the experiments. Histological examination revealed that the FGF1 coacervate reduced inflammation and fibrosis post-MI, significantly increased the proliferation of endothelial and mural cells, and resulted in stable arterioles and capillaries. Furthermore, the FGF1 coacervate improved the proliferation of cardiac stem cells 6 weeks post-MI. However, free FGF1, dosed identically, did not show significant difference from saline treatment. Thus, one injection of FGF1 coacervate was sufficient to attenuate the injury caused by MI, and the results were significantly better than those obtained from an equal dose of free FGF1.
Guarda su PubMed -
3D Printed Stem-Cell-Laden, Microchanneled Hydrogel Patch for the Enhanced Release of Cell-Secreting Factors and Treatment of Myocardial Infarctions.
ACS Biomater Sci Eng2017 Sep;3(9):1980-1987. doi: 10.1021/acsbiomaterials.6b00176.
Melhem Molly R, Park Jooyeon, Knapp Luke, Reinkensmeyer Larissa, Cvetkovic Caroline, Flewellyn Jordan, Lee Min Kyung, Jensen Tor Wolf, Bashir Rashid, Kong Hyunjoon, Schook Lawrence B,
Abstract
Over the past several years, biomaterials loaded with mesenchymal stem cells (MSCs) have increasingly been used to reduce the myocardial fate of postinfarction collagen deposition and scar tissue formation. Despite successful gains, therapeutic efficacy has remained limited because of restricted transport of cell-secreting factors at the site of implantation. We hypothesized that an MSC-laden hydrogel patch with multiple microchannels would retain transplanted cells on target tissue and support transport of cell-secreting factors into tissue. By doing so, the gel patch will improve the therapeutic potential of the cells and minimize the degradation of myocardial tissue postinfarction. To examine this hypothesis, a stereolithographic apparatus (SLA) was used to introduce microchannels of controlled diameters (e.g., 500 and 1000 ?m) during in situ cross-linking reaction of poly(ethylene glycol)dimethacrylate solution suspended with cells. Placement of the MSC-laden, microchanneled gel patch on the occluded left coronary artery in a murine model showed significant improvement in the ejection fraction, fractional shortening, and stroke volume, compared with gel patches without MSCs and MSC-laden gel patches without microchannels. In particular, the microchannels significantly reduced the number of cells required to recover cardiac function, while minimizing cardiac remodeling. In sum, the microchanneled gel patch would provide a means to prevent abnormal fibrosis resulting from acute ischemic injury.
Guarda su PubMed -
Oxygen-Generating Photo-Cross-Linkable Hydrogels Support Cardiac Progenitor Cell Survival by Reducing Hypoxia-Induced Necrosis.
ACS Biomater Sci Eng2017 Sep;3(9):1964-1971. doi: 10.1021/acsbiomaterials.6b00109.
Alemdar Neslihan, Leijten Jeroen, Camci-Unal Gulden, Hjortnaes Jesper, Ribas Joao, Paul Arghya, Mostafalu Pooria, Gaharwar Akhilesh K, Qiu Yiling, Sonkusale Sameer, Liao Ronglih, Khademhosseini Ali,
Abstract
Oxygen is essential to cell survival and tissue function. Not surprisingly, ischemia resulting from myocardial infarction induces cell death and tissue necrosis. Attempts to regenerate myocardial tissue with cell based therapies exacerbate the hypoxic stress by further increasing the metabolic burden. In consequence, implanted tissue engineered cardiac tissues suffer from hypoxia-induced cell death. Here, we report on the generation of oxygen-generating hydrogels composed of calcium peroxide (CPO) laden gelatin methacryloyl (GelMA). CPO-GelMA hydrogels released significant amounts of oxygen for over a period of 5 days under hypoxic conditions (1% O). The released oxygen proved sufficient to relieve the metabolic stress of cardiac side population cells that were encapsulated within CPO-GelMA hydrogels. In particular, incorporation of CPO in GelMA hydrogels strongly enhanced cell viability as compared to GelMA-only hydrogels. Importantly, CPO-based oxygen generation reduced cell death by limiting hypoxia-induced necrosis. The current study demonstrates that CPO based oxygen-generating hydrogels could be used to transiently provide oxygen to cardiac cells under ischemic conditions. Therefore, oxygen generating materials such as CPO-GelMA can improve cell-based therapies aimed at treatment or regeneration of infarcted myocardial tissue.
Guarda su PubMed -
Human Stem Cell-Derived Cardiac Model of Chronic Drug Exposure.
ACS Biomater Sci Eng2017 Sep;3(9):1911-1921. doi: 10.1021/acsbiomaterials.5b00496.
Nunes Sara S, Feric Nicole, Pahnke Aric, Miklas Jason W, Li Mark, Coles John, Gagliardi Mark, Keller Gordon, Radisic Milica,
Abstract
Animal models have been instrumental in providing insight into the molecular basis of disease. While such information has been successfully applied to the study of human disease, this translation would be significantly strengthened by the availability of models based on human cells. This would be particularly important for cardiovascular disease, as the physiology of human cardiomyocytes (CMs) differs significantly from rodents. Here, we have generated a three-dimensional human engineered cardiac tissue, termed biowire, from human embryonic stem cell-derived CMs to investigate the effects of chronic (7 day) treatment with isoproterenol, endothelin-1, or angiotensin II. We show that biowires chronically treated with either isoproterenol, endothelin-1, or angiotensin II have disrupted myofibril alignment and significantly reduced force of contraction. Isoproterenol-treated biowires have upregulated brain natriuretic peptide and atrial natriuretic peptide gene expression. Endothelin-1 and angiotensin II-treated biowires demonstrated a significantly increased cell size. Endothelin-1-treated biowires exhibited increased cardiac troponin secretion into the culture media. This demonstrates that human biowires treated for 7 days with isoproterenol, angiotensin II, or endothelin-1 exhibit some changes compatible with hypertrophic cardiomyopathy.
Guarda su PubMed -
Shortcomings of Animal Models and the Rise of Engineered Human Cardiac Tissue.
ACS Biomater Sci Eng2017 Sep;3(9):1884-1897. doi: 10.1021/acsbiomaterials.6b00662.
Fine Barry, Vunjak-Novakovic Gordana,
Abstract
We provide here an historical context of how studies utilizing engineered human cardiac muscle can complement and in some cases substitute animal and cell models for studies of disease and drug testing. We give an overview of the development of animal models and discuss the ability of novel human tissue models to overcome limited predictive power of cell culture and animal models in studies of drug efficacy and safety. The in vitro generation of cardiac tissue is discussed in the context of state of the art in the field. Finally we describe the assembly of multitissue platforms for more accurate representation of integrated human cardiac physiology and consider the advantages of in silico drug trials to augment our ability to predict drug-drug and organ-organ interactions in humans.
Guarda su PubMed
