Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 16.078
Filtrar
1.
Bull Exp Biol Med ; 177(1): 115-123, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38963596

RESUMO

The cardiac perivascular niche is a cellular microenvironment of a blood vessel. The principles of niche regulation are still poorly understood. We studied the effect of TGFß1 on cells forming the cardiac perivascular niche using 3D cell culture (cardiospheres). Cardiospheres contained progenitor (c-Kit), endothelial (CD31), and mural (αSMA) cells, basement membrane proteins (laminin) and extracellular matrix proteins (collagen I, fibronectin). TGFß1 treatment decreased the length of CD31+ microvasculature, VE cadherin protein level, and proportion of NG2+ cells, and increased proportion of αSMA+ cells and transgelin/SM22α protein level. We supposed that this effect is related to the stabilizing function of TGFß1 on vascular cells: decreased endothelial cell proliferation, as shown for HUVEC, and activation of mural cell differentiation.


Assuntos
Diferenciação Celular , Proliferação de Células , Fator de Crescimento Transformador beta1 , Fator de Crescimento Transformador beta1/farmacologia , Fator de Crescimento Transformador beta1/metabolismo , Diferenciação Celular/efeitos dos fármacos , Humanos , Proliferação de Células/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Animais , Proteínas dos Microfilamentos/metabolismo , Proteínas dos Microfilamentos/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Caderinas/metabolismo , Laminina/metabolismo , Laminina/farmacologia , Proteínas Musculares/metabolismo , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/citologia , Fibronectinas/metabolismo , Fibronectinas/farmacologia , Antígenos CD/metabolismo , Miocárdio/metabolismo , Miocárdio/citologia , Nicho de Células-Tronco/efeitos dos fármacos , Nicho de Células-Tronco/fisiologia , Colágeno Tipo I/metabolismo , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/citologia , Técnicas de Cultura de Células em Três Dimensões/métodos
2.
Curr Protoc ; 4(7): e1097, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39036931

RESUMO

In the heart in vivo, vasculature forms a semi-permeable endothelial barrier for selective nutrient and (immune) cell delivery to the myocardium and removal of waste products. Crosstalk between the vasculature and the heart cells regulates homeostasis in health and disease. To model heart development and disease in vitro it is important that essential features of this crosstalk are captured. Cardiac organoid and microtissue models often integrate endothelial cells (ECs) to form microvascular networks inside the 3D structure. However, in static culture without perfusion, these networks may fail to show essential functionality. Here, we describe a protocol to generate an in vitro model of human induced pluripotent stem cell (hiPSC)-derived vascularized cardiac microtissues on a microfluidic organ-on-chip platform (VMToC) in which the blood vessels are perfusable. First, prevascularized cardiac microtissues (MT) are formed by combining hiPSC-derived cardiomyocytes, ECs, and cardiac fibroblasts in a pre-defined ratio. Next, these prevascularized MTs are integrated in the chips in a fibrin hydrogel containing additional vascular cells, which self-organize into tubular structures. The MTs become vascularized through anastomosis between the pre-existing microvasculature in the MT and the external vascular network. The VMToCs are then ready for downstream structural and functional assays and basic characterization. Using this protocol, cardiac MTs can be efficiently and robustly vascularized and perfused within 7 days. In vitro vascularized organoid and MT models have the potential to transition current 3D cardiac models to more physiologically relevant organ models that allow the role of the endothelial barrier in drug and inflammatory response to be investigated. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol: Generation of VMToC Support Protocol 1: Functional Characterization of VMToC Support Protocol 2: Structural Characterization of VMToC.


Assuntos
Células-Tronco Pluripotentes Induzidas , Dispositivos Lab-On-A-Chip , Miócitos Cardíacos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/citologia , Células Endoteliais/citologia , Miocárdio/citologia , Miocárdio/metabolismo , Diferenciação Celular
3.
Sci Rep ; 14(1): 13085, 2024 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-38849466

RESUMO

The response of cardiac fibroblast proliferation to detrimental stimuli is one of the main pathological factors causing heart remodeling. Reactive oxygen species (ROS) mediate the proliferation of cardiac fibroblasts. However, the exact molecular mechanism remains unclear. In vivo, we examined the oxidative modification of miRNAs with miRNA immunoprecipitation with O8G in animal models of cardiac fibrosis induced by Ang II injection or ischemia‒reperfusion injury. Furthermore, in vitro, we constructed oxidation-modified miR-30c and investigated its effects on the proliferation of cardiac fibroblasts. Additionally, luciferase reporter assays were used to identify the target of oxidized miR-30c. We found that miR-30c oxidation was modified by Ang II and PDGF treatment and mediated by excess ROS. We demonstrated that oxidative modification of G to O8G occurred at positions 4 and 5 of the 5' end of miR-30c (4,5-oxo-miR-30c), and this modification promoted cardiac fibroblast proliferation. Furthermore, CDKN2C is a negative regulator of cardiac fibroblast proliferation. 4,5-oxo-miR-30c misrecognizes CDKN2C mRNA, resulting in a reduction in protein expression. Oxidized miR-30c promotes cardiac fibroblast proliferation by mismatch mRNA of CDKN2C.


Assuntos
Proliferação de Células , Fibroblastos , MicroRNAs , Oxirredução , MicroRNAs/genética , MicroRNAs/metabolismo , Animais , Fibroblastos/metabolismo , Fibroblastos/citologia , Espécies Reativas de Oxigênio/metabolismo , Miocárdio/metabolismo , Miocárdio/citologia , Angiotensina II/farmacologia , Ratos , Masculino , Camundongos , Fibrose
4.
Circ Res ; 134(12): 1791-1807, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38843293

RESUMO

Cardiac macrophages represent a functionally diverse population of cells involved in cardiac homeostasis, repair, and remodeling. With recent advancements in single-cell technologies, it is possible to elucidate specific macrophage subsets based on transcriptional signatures and cell surface protein expression to gain a deep understanding of macrophage diversity in the heart. The use of fate-mapping technologies and parabiosis studies have provided insight into the ontogeny and dynamics of macrophages identifying subsets derived from embryonic and adult definitive hematopoietic progenitors that include tissue-resident and bone marrow monocyte-derived macrophages, respectively. Within the heart, these subsets have distinct tissue niches and functional roles in the setting of homeostasis and disease, with cardiac resident macrophages representing a protective cell population while bone marrow monocyte-derived cardiac macrophages have a context-dependent effect, triggering both proinflammatory tissue injury, but also promoting reparative functions. With the increased understanding of the clinical relevance of cardiac macrophage subsets, there has been an increasing need to detect and measure cardiac macrophage compositions in living animals and patients. New molecular tracers compatible with positron emission tomography/computerized tomography and positron emission tomography/ magnetic resonance imaging have enabled investigators to noninvasively and serially visualize cardiac macrophage subsets within the heart to define associations with disease and measure treatment responses. Today, advancements within this thriving field are poised to fuel an era of clinical translation.


Assuntos
Macrófagos , Miocárdio , Animais , Macrófagos/metabolismo , Humanos , Miocárdio/metabolismo , Miocárdio/citologia
5.
Physiol Rep ; 12(11): e16108, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38872461

RESUMO

ERK3/MAPK6 activates MAP kinase-activated protein kinase (MK)-5 in selected cell types. Male MK5 haplodeficient mice show reduced hypertrophy and attenuated increase in Col1a1 mRNA in response to increased cardiac afterload. In addition, MK5 deficiency impairs cardiac fibroblast function. This study determined the effect of reduced ERK3 on cardiac hypertrophy following transverse aortic constriction (TAC) and fibroblast biology in male mice. Three weeks post-surgery, ERK3, but not ERK4 or p38α, co-immunoprecipitated with MK5 from both sham and TAC heart lysates. The increase in left ventricular mass and myocyte diameter was lower in TAC-ERK3+/- than TAC-ERK3+/+ hearts, whereas ERK3 haploinsufficiency did not alter systolic or diastolic function. Furthermore, the TAC-induced increase in Col1a1 mRNA abundance was diminished in ERK3+/- hearts. ERK3 immunoreactivity was detected in atrial and ventricular fibroblasts but not myocytes. In both quiescent fibroblasts and "activated" myofibroblasts isolated from adult mouse heart, siRNA-mediated knockdown of ERK3 reduced the TGF-ß-induced increase in Col1a1 mRNA. In addition, intracellular type 1 collagen immunoreactivity was reduced following ERK3 depletion in quiescent fibroblasts but not myofibroblasts. Finally, knocking down ERK3 impaired motility in both atrial and ventricular myofibroblasts. These results suggest that ERK3 plays an important role in multiple aspects of cardiac fibroblast biology.


Assuntos
Fibroblastos , Animais , Masculino , Camundongos , Fibroblastos/metabolismo , Colágeno Tipo I/metabolismo , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I/metabolismo , Miocárdio/metabolismo , Miocárdio/citologia , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Proteína Quinase 6 Ativada por Mitógeno/genética , Camundongos Endogâmicos C57BL , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Células Cultivadas , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Cardiomegalia/genética , Miócitos Cardíacos/metabolismo
6.
Cell Biochem Funct ; 42(4): e4066, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38822669

RESUMO

Collagen crosslinking, mediated by lysyl oxidase, is an adaptive mechanism of the cardiac repair process initiated by cardiac fibroblasts postmyocardial injury. However, excessive crosslinking leads to cardiac wall stiffening, which impairs the contractile properties of the left ventricle and leads to heart failure. In this study, we investigated the role of periostin, a matricellular protein, in the regulation of lysyl oxidase in cardiac fibroblasts in response to angiotensin II and TGFß1. Our results indicated that periostin silencing abolished the angiotensin II and TGFß1-mediated upregulation of lysyl oxidase. Furthermore, the attenuation of periostin expression resulted in a notable reduction in the activity of lysyl oxidase. Downstream of periostin, ERK1/2 MAPK signaling was found to be activated, which in turn transcriptionally upregulates the serum response factor to facilitate the enhanced expression of lysyl oxidase. The periostin-lysyl oxidase association was also positively correlated in an in vivo rat model of myocardial infarction. The expression of periostin and lysyl oxidase was upregulated in the collagen-rich fibrotic scar tissue of the left ventricle. Remarkably, echocardiography data showed a reduction in the left ventricular wall movement, ejection fraction, and fractional shortening, indicative of enhanced stiffening of the cardiac wall. These findings shed light on the mechanistic role of periostin in the collagen crosslinking initiated by activated cardiac fibroblasts. Our findings signify periostin as a possible therapeutic target to reduce excessive collagen crosslinking that contributes to the structural remodeling associated with heart failure.


Assuntos
Moléculas de Adesão Celular , Fibroblastos , Proteína-Lisina 6-Oxidase , Ratos Sprague-Dawley , Animais , Proteína-Lisina 6-Oxidase/metabolismo , Fibroblastos/metabolismo , Ratos , Moléculas de Adesão Celular/metabolismo , Masculino , Sistema de Sinalização das MAP Quinases , Miocárdio/metabolismo , Miocárdio/citologia , Angiotensina II/farmacologia , Angiotensina II/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Células Cultivadas , Modelos Animais de Doenças , Periostina
7.
Adv Exp Med Biol ; 1441: 103-124, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38884707

RESUMO

The heart forms from the first and second heart fields, which contribute to distinct regions of the myocardium. This is supported by clonal analyses, which identify corresponding first and second cardiac cell lineages in the heart. Progenitor cells of the second heart field and its sub-domains are controlled by a gene regulatory network and signaling pathways, which determine their behavior. Multipotent cells in this field can also contribute cardiac endothelial and smooth muscle cells. Furthermore, the skeletal muscles of the head and neck are clonally related to myocardial cells that form the arterial and venous poles of the heart. These lineage relationships, together with the genes that regulate the heart fields, have major implications for congenital heart disease.


Assuntos
Linhagem da Célula , Animais , Humanos , Diferenciação Celular/genética , Linhagem da Célula/genética , Coração/fisiologia , Miocárdio/citologia , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Transdução de Sinais , Células-Tronco/metabolismo , Células-Tronco/citologia , Células-Tronco/fisiologia
8.
Sci Rep ; 14(1): 14329, 2024 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-38907041

RESUMO

A comprehensive grasp of the myocardial micro-architecture is essential for understanding diverse heart functions. This study aimed to investigate three-dimensional (3D) cardiomyocyte arrangement in the laminar structure using X-ray phase-contrast microtomography. Using the ID-19 beamline at the European Synchrotron Radiation Facility, we imaged human left ventricular (LV) wall transparietal samples and reconstructed them with an isotropic voxel edge length of 3.5 µm. From the reconstructed volumes, we extracted different regions to analyze the orientation distribution of local cardiomyocyte aggregates, presenting findings in terms of helix and intrusion angles. In regions containing one sheetlet population, we observed cardiomyocyte aggregates running along the local LV wall's radial direction at the border of sheetlets, branching and merging into a complex network around connecting points of different sheetlets, and bending to accommodate vessel passages. In regions with two sheetlet populations, the helix angle of local cardiomyocyte aggregates experiences a nonmonotonic change, and some cardiomyocyte aggregates run along the local radial direction. X-ray phase-contrast microtomography is a valuable technique for investigating the 3D local myocardial architecture at microscopic level. The arrangement of local cardiomyocyte aggregates in the LV wall proves to be both regional and complex, intricately linked to the local laminar structure.


Assuntos
Ventrículos do Coração , Imageamento Tridimensional , Miócitos Cardíacos , Microtomografia por Raio-X , Microtomografia por Raio-X/métodos , Humanos , Miócitos Cardíacos/citologia , Imageamento Tridimensional/métodos , Ventrículos do Coração/diagnóstico por imagem , Miocárdio/citologia
9.
Cell Rep ; 43(6): 114302, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38824644

RESUMO

Resident cardiac macrophages are critical mediators of cardiac function. Despite their known importance to cardiac electrophysiology and tissue maintenance, there are currently no stem-cell-derived models of human engineered cardiac tissues (hECTs) that include resident macrophages. In this study, we made an induced pluripotent stem cell (iPSC)-derived hECT model with a resident population of macrophages (iM0) to better recapitulate the native myocardium and characterized their impact on tissue function. Macrophage retention within the hECTs was confirmed via immunofluorescence after 28 days of cultivation. The inclusion of iM0s significantly impacted hECT function, increasing contractile force production. A potential mechanism underlying these changes was revealed by the interrogation of calcium signaling, which demonstrated the modulation of ß-adrenergic signaling in +iM0 hECTs. Collectively, these findings demonstrate that macrophages significantly enhance cardiac function in iPSC-derived hECT models, emphasizing the need to further explore their contributions not only in healthy hECT models but also in the contexts of disease and injury.


Assuntos
Células-Tronco Pluripotentes Induzidas , Macrófagos , Contração Miocárdica , Engenharia Tecidual , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Macrófagos/metabolismo , Engenharia Tecidual/métodos , Contração Miocárdica/fisiologia , Miocárdio/metabolismo , Miocárdio/citologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/citologia , Diferenciação Celular , Sinalização do Cálcio
10.
Cell Rep Methods ; 4(6): 100798, 2024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38889687

RESUMO

Stem cell organoids are powerful models for studying organ development, disease modeling, drug screening, and regenerative medicine applications. The convergence of organoid technology, tissue engineering, and artificial intelligence (AI) could potentially enhance our understanding of the design principles for organoid engineering. In this study, we utilized micropatterning techniques to create a designer library of 230 cardiac organoids with 7 geometric designs. We employed manifold learning techniques to analyze single organoid heterogeneity based on 10 physiological parameters. We clustered and refined the cardiac organoids based on their functional similarity using unsupervised machine learning approaches, thus elucidating unique functionalities associated with geometric designs. We also highlighted the critical role of calcium transient rising time in distinguishing organoids based on geometric patterns and clustering results. This integration of organoid engineering and machine learning enhances our understanding of structure-function relationships in cardiac organoids, paving the way for more controlled and optimized organoid design.


Assuntos
Aprendizado de Máquina , Organoides , Engenharia Tecidual , Organoides/citologia , Engenharia Tecidual/métodos , Humanos , Animais , Coração/fisiologia , Miocárdio/citologia , Miocárdio/metabolismo
11.
Int J Mol Sci ; 25(12)2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38928463

RESUMO

The prevalence of dilated cardiomyopathy (DCM) is increasing globally, highlighting the need for innovative therapeutic approaches to prevent its onset. In this study, we examined the energetic and epigenetic distinctions between dilated and non-dilated human myocardium-derived mesenchymal stem/stromal cells (hmMSCs) and assessed the effects of class I and II HDAC inhibitors (HDACi) on these cells and their cardiomyogenic differentiation. Cells were isolated from myocardium biopsies using explant outgrowth methods. Mitochondrial and histone deacetylase activities, ATP levels, cardiac transcription factors, and structural proteins were assessed using flow cytometry, PCR, chemiluminescence, Western blotting, and immunohistochemistry. The data suggest that the tested HDAC inhibitors improved acetylation and enhanced the energetic status of both types of cells, with significant effects observed in dilated myocardium-derived hmMSCs. Additionally, the HDAC inhibitors activated the cardiac transcription factors Nkx2-5, HOPX, GATA4, and Mef2C, and upregulated structural proteins such as cardiac troponin T and alpha cardiac actin at both the protein and gene levels. In conclusion, our findings suggest that HDACi may serve as potential modulators of the energetic status and cardiomyogenic differentiation of human heart hmMSCs. This avenue of exploration could broaden the search for novel therapeutic interventions for dilated cardiomyopathy, ultimately leading to improvements in heart function.


Assuntos
Cardiomiopatia Dilatada , Diferenciação Celular , Inibidores de Histona Desacetilases , Células-Tronco Mesenquimais , Humanos , Inibidores de Histona Desacetilases/farmacologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Diferenciação Celular/efeitos dos fármacos , Miocárdio/citologia , Miocárdio/metabolismo , Miocárdio/patologia , Histona Desacetilases/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/citologia , Fatores de Transcrição MEF2/metabolismo , Fatores de Transcrição MEF2/genética , Proteína Homeobox Nkx-2.5/metabolismo , Proteína Homeobox Nkx-2.5/genética , Acetilação/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Células Cultivadas
13.
Biochem Soc Trans ; 52(3): 1339-1348, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38775188

RESUMO

Myocardial cell fate specification takes place during the early stages of heart development as the precardiac mesoderm is configured into two symmetrical sets of bilateral precursor cells. Molecular cues of the surrounding tissues specify and subsequently determine the early cardiomyocytes, that finally matured as the heart is completed at early postnatal stages. Over the last decade, we have greatly enhanced our understanding of the transcriptional regulation of cardiac development and thus of myocardial cell fate. The recent discovery of a novel layer of gene regulation by non-coding RNAs has flourished their implication in epigenetic, transcriptional and post-transcriptional regulation of cardiac development. In this review, we revised the current state-of-the-art knowledge on the functional role of non-coding RNAs during myocardial cell fate.


Assuntos
Diferenciação Celular , Miócitos Cardíacos , RNA não Traduzido , Humanos , Animais , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/citologia , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Miocárdio/metabolismo , Miocárdio/citologia , Coração/embriologia , Epigênese Genética , Linhagem da Célula
15.
Comput Biol Med ; 177: 108624, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38795420

RESUMO

BACKGROUND: Analysis of structures contained in tissue samples and the relevant contextual information is of utmost importance to histopathologists during diagnosis. Cardiac biopsies require in-depth analysis of the relationships between biological structures. Statistical measures are insufficient for determining a model's viability and applicability in the diagnostic process. A deeper understanding of predictions is necessary in order to support histopathologists. METHODS: We propose a method for providing supporting information in the form of segmentation of histological structures to histopathologists based on these principles. The proposed method utilizes nuclei type and density information in addition to standard image input provided at two different zoom levels for the semantic segmentation of blood vessels, inflammation, and endocardium in heart tissue. RESULTS: The proposed method was able to reach state-of-the-art segmentation results. The overall quality and viability of the predictions was qualitatively evaluated by two pathologists and a histotechnologist. CONCLUSIONS: The decision process of the proposed deep learning model utilizes the provided information sources correctly and simulates the decision process of histopathologists via the usage of a custom-designed attention gate that provides a combination of spatial and encoder attention mechanisms. The implementation is available at https://github.com/mathali/IEDL-segmentation-of-heart-tissue.


Assuntos
Aprendizado Profundo , Humanos , Miocárdio/patologia , Miocárdio/citologia , Semântica , Processamento de Imagem Assistida por Computador/métodos , Coração/diagnóstico por imagem , Coração/anatomia & histologia
17.
Biomed Mater ; 19(4)2024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38815609

RESUMO

The alignment of each cell in human myocardium is considered critical for the efficient movement of cardiac tissue. We investigated 96-well microstripe-patterned plates to align human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs), which resemble fetal myocardium. The aligned CMs (ACMs) cultured on the microstripe-patterned plates exhibited pathology, motor function, gene expression, and drug response that more closely resembled those of adult cells than did unaligned CMs cultured on a flat plate (FCMs). We used these ACMs to evaluate drug side effects and efficacy, and to determine whether these were similar to adult-like responses. When CMs from patients with hypertrophic cardiomyopathy (HCMs) were seeded and cultured on the microstripe-patterned plates or layered on top of the ACMs, both sets of HCMs showed increased heart rate and synchronized contractions, indicating improved cardiac function. It is suggested that the ACMs could be used for drug screening as cells representative of adult-like CMs and be transplanted in the form of a cell sheet for regenerative treatment of heart failure.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas , Miócitos Cardíacos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Cardiomiopatia Hipertrófica , Células Cultivadas , Miocárdio/citologia , Miocárdio/metabolismo , Engenharia Tecidual/métodos , Técnicas de Cultura de Células
18.
Sci Rep ; 14(1): 10365, 2024 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-38710778

RESUMO

Cardiac fibroblasts (CFs) are essential for preserving myocardial integrity and function. They can detect variations in cardiac tissue stiffness using various cellular mechanosensors, including the Ca2+ permeable mechanosensitive channel Piezo1. Nevertheless, how CFs adapt the mechanosensitive response to stiffness changes remains unclear. In this work we adopted a multimodal approach, combining the local mechanical stimulation (from 10 pN to 350 nN) with variations of culture substrate stiffness. We found that primary rat CFs cultured on stiff (GPa) substrates showed a broad Piezo1 distribution in the cell with particular accumulation at the mitochondria membrane. CFs displayed a force-dependent behavior in both calcium uptake and channel activation probability, showing a threshold at 300 nN, which involves both cytosolic and mitochondrial Ca2+ mobilization. This trend decreases as the myofibroblast phenotype within the cell population increases, following a possible Piezo1 accumulation at focal adhesion sites. In contrast, the inhibition of fibroblasts to myofibroblasts transition with soft substrates (kPa) considerably reduces both mechanically- and chemically-induced Piezo1 activation and expression. Our findings shed light on how Piezo1 function and expression are regulated by the substrate stiffness and highlight its involvement in the environment-mediated modulation of CFs mechanosensitivity.


Assuntos
Fibroblastos , Canais Iônicos , Mecanotransdução Celular , Proteínas de Membrana , Animais , Canais Iônicos/metabolismo , Ratos , Fibroblastos/metabolismo , Fibroblastos/citologia , Células Cultivadas , Cálcio/metabolismo , Miofibroblastos/metabolismo , Miofibroblastos/fisiologia , Miocárdio/metabolismo , Miocárdio/citologia , Microambiente Celular
19.
J Mech Behav Biomed Mater ; 155: 106571, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38744118

RESUMO

Cardiac fibrosis refers to the abnormal accumulation of extracellular matrix within the cardiac muscle, leading to increased stiffness and impaired heart function. From a rheological standpoint, knowledge about myocardial behavior is still lacking, partially due to a lack of appropriate techniques to investigate the rheology of in vitro cardiac tissue models. 3D multicellular cardiac spheroids are powerful and versatile platforms for modeling healthy and fibrotic cardiac tissue in vitro and studying how their mechanical properties are modulated. In this study, cardiac spheroids were created by co-culturing neonatal rat ventricular cardiomyocytes and fibroblasts in definite ratios using the hanging-drop method. The rheological characterization of such models was performed by Atomic Force Microscopy-based stress-relaxation measurements on the whole spheroid. After strain application, a viscoelastic bi-exponential relaxation was observed, characterized by a fast relaxation time (τ1) followed by a slower one (τ2). In particular, spheroids with higher fibroblasts density showed reduction for both relaxation times comparing to control, with a more pronounced decrement of τ1 with respect to τ2. Such response was found compatible with the increased production of extracellular matrix within these spheroids, which recapitulates the main feature of the fibrosis pathophysiology. These results demonstrate how the rheological characteristics of cardiac tissue vary as a function of cellular composition and extracellular matrix, confirming the suitability of such system as an in vitro preclinical model of cardiac fibrosis.


Assuntos
Fibrose , Miócitos Cardíacos , Reologia , Esferoides Celulares , Animais , Esferoides Celulares/citologia , Esferoides Celulares/patologia , Ratos , Miócitos Cardíacos/citologia , Fibroblastos/citologia , Miocárdio/citologia , Miocárdio/patologia , Miocárdio/metabolismo , Ratos Wistar , Modelos Biológicos
20.
BMC Med Ethics ; 25(1): 61, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38773517

RESUMO

Certain organoid subtypes are particularly sensitive. We explore whether moral intuitions about the heartbeat warrant unique moral consideration for newly advanced contracting cardiac organoids. Despite the heartbeat's moral significance in organ procurement and abortion discussions, we argue that this significance should not translate into moral implications for cardiac organoids.


Assuntos
Princípios Morais , Organoides , Humanos , Obtenção de Tecidos e Órgãos/ética , Coração/fisiologia , Miocárdio/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...