Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biomater Sci ; 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38910521

RESUMEN

Cardiac tissue engineering (cTE) has already advanced towards the first clinical trials, investigating safety and feasibility of cTE construct transplantation in failing hearts. However, the lack of well-established preservation methods poses a hindrance to further scalability, commercialization, and transportation, thereby reducing their clinical implementation. In this study, hypothermic preservation (4 °C) and two methods for cryopreservation (i.e., a slow and fast cooling approach to -196 °C and -150 °C, respectively) were investigated as potential solutions to extend the cTE construct implantation window. The cTE model used consisted of human induced pluripotent stem cell-derived cardiomyocytes and human cardiac fibroblasts embedded in a natural-derived hydrogel and supported by a polymeric melt electrowritten hexagonal scaffold. Constructs, composed of cardiomyocytes of different maturity, were preserved for three days, using several commercially available preservation protocols and solutions. Cardiomyocyte viability, function (beat rate and calcium handling), and metabolic activity were investigated after rewarming. Our observations show that cardiomyocytes' age did not influence post-rewarming viability, however, it influenced construct function. Hypothermic preservation with HypoThermosol® ensured cardiomyocyte viability and function. Furthermore, fast freezing outperformed slow freezing, but both viability and function were severely reduced after rewarming. In conclusion, whereas long-term preservation remains a challenge, hypothermic preservation with HypoThermosol® represents a promising solution for cTE construct short-term preservation and potential transportation, aiding in off-the-shelf availability, ultimately increasing their clinical applicability.

3.
J Vis Exp ; (193)2023 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-36971448

RESUMEN

Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are of paramount importance for human cardiac disease modeling and therapeutics. We recently published a cost-effective strategy for the massive expansion of hiPSC-CMs in two dimensions (2D). Two major limitations are cell immaturity and a lack of three-dimensional (3D) arrangement and scalability in high-throughput screening (HTS) platforms. To overcome these limitations, the expanded cardiomyocytes form an ideal cell source for the generation of 3D cardiac cell culture and tissue engineering techniques. The latter holds great potential in the cardiovascular field, providing more advanced and physiologically relevant HTS. Here, we describe an HTS-compatible workflow with easy scalability for the generation, maintenance, and optical analysis of cardiac spheroids (CSs) in a 96-well-format. These small CSs are essential to fill the gap present in current in vitro disease models and/or generation for 3D tissue engineering platforms. The CSs present a highly structured morphology, size, and cellular composition. Furthermore, hiPSC-CMs cultured as CSs display increased maturation and several functional features of the human heart, such as spontaneous calcium handling and contractile activity. By automatization of the complete workflow, from the generation of CSs to functional analysis, we increase intra- and inter-batch reproducibility as demonstrated by high-throughput (HT) imaging and calcium handling analysis. The described protocol allows modeling of cardiac diseases and assessing drug/therapeutic effects at the single-cell level within a complex 3D cell environment in a fully automated HTS workflow. In addition, the study describes a straightforward procedure for long-term preservation and biobanking of whole-spheroids, thereby providing researchers the opportunity to create next-generation functional tissue storage. HTS combined with long-term storage will substantially contribute to translational research in a wide range of areas, including drug discovery and testing, regenerative medicine, and the development of personalized therapies.


Asunto(s)
Cardiopatías , Células Madre Pluripotentes Inducidas , Humanos , Ensayos Analíticos de Alto Rendimiento , Calcio/farmacología , Bancos de Muestras Biológicas , Reproducibilidad de los Resultados , Miocitos Cardíacos , Diferenciación Celular/fisiología
5.
Front Pharmacol ; 13: 869512, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35694249

RESUMEN

Circadian rhythms influence the recruitment of immune cells and the onset of inflammation, which is pivotal in the response to ischemic cardiac injury after a myocardial infarction (MI). The hyperacute immune response that occurs within the first few hours after a MI has not yet been elucidated. Therefore, we characterized the immune response and myocardial damage 3 hours after a MI occurs over a full twenty-four-hour period to investigate the role of the circadian rhythms in this response. MI was induced at Zeitgeber Time (ZT) 2, 8, 14, and 20 by permanent ligation of the left anterior descending coronary artery. Three hours after surgery, animals were terminated and blood and hearts collected to assess the immunological status and cardiac damage. Blood leukocyte numbers varied throughout the day, peaking during the rest-phase (ZT2 and 8). Extravasation of leukocytes was more pronounced during the active-phase (ZT14 and 20) and was associated with greater chemokine release to the blood and expression of adhesion molecules in the heart. Damage to the heart, measured by Troponin-I plasma levels, was elevated during this time frame. Clock gene oscillations remained intact in both MI-induced and sham-operated mice hearts, which could explain the circadian influence of the hyperacute inflammatory response after a MI. These findings are in line with the clinical observation that patients who experience a MI early in the morning (i.e., early active phase) have worse clinical outcomes. This study provides further insight on the immune response occurring shortly after an MI, which may contribute to the development of novel and optimization of current therapeutic approaches.

6.
Mol Ther ; 30(6): 2257-2273, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35278675

RESUMEN

As mediators of intercellular communication, extracellular vesicles containing molecular cargo, such as microRNAs, are secreted by cells and taken up by recipient cells to influence their cellular phenotype and function. Here we report that cardiac stress-induced differential microRNA content, with miR-200c-3p being one of the most enriched, in cardiomyocyte-derived extracellular vesicles mediates functional cross-talk with endothelial cells. Silencing of miR-200c-3p in mice subjected to chronic increased cardiac pressure overload resulted in attenuated hypertrophy, smaller fibrotic areas, higher capillary density, and preserved cardiac ejection fraction. We were able to maximally rescue microvascular and cardiac function with very low doses of antagomir, which specifically silences miR-200c-3p expression in non-myocyte cells. Our results reveal vesicle transfer of miR-200c-3p from cardiomyocytes to cardiac endothelial cells, underlining the importance of cardiac intercellular communication in the pathophysiology of heart failure.


Asunto(s)
Vesículas Extracelulares , MicroARNs , Animales , Comunicación Celular , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo
7.
Nat Commun ; 12(1): 4808, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34376683

RESUMEN

Myocardial regeneration is restricted to early postnatal life, when mammalian cardiomyocytes still retain the ability to proliferate. The molecular cues that induce cell cycle arrest of neonatal cardiomyocytes towards terminally differentiated adult heart muscle cells remain obscure. Here we report that the miR-106b~25 cluster is higher expressed in the early postnatal myocardium and decreases in expression towards adulthood, especially under conditions of overload, and orchestrates the transition of cardiomyocyte hyperplasia towards cell cycle arrest and hypertrophy by virtue of its targetome. In line, gene delivery of miR-106b~25 to the mouse heart provokes cardiomyocyte proliferation by targeting a network of negative cell cycle regulators including E2f5, Cdkn1c, Ccne1 and Wee1. Conversely, gene-targeted miR-106b~25 null mice display spontaneous hypertrophic remodeling and exaggerated remodeling to overload by derepression of the prohypertrophic transcription factors Hand2 and Mef2d. Taking advantage of the regulatory function of miR-106b~25 on cardiomyocyte hyperplasia and hypertrophy, viral gene delivery of miR-106b~25 provokes nearly complete regeneration of the adult myocardium after ischemic injury. Our data demonstrate that exploitation of conserved molecular programs can enhance the regenerative capacity of the injured heart.


Asunto(s)
MicroARNs/genética , Infarto del Miocardio/genética , Miocitos Cardíacos/metabolismo , Regeneración/genética , Animales , Animales Recién Nacidos , Cardiomegalia/genética , Células Cultivadas , Ecocardiografía , Regulación de la Expresión Génica , Humanos , Hiperplasia/genética , Ratones , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Adv Mater ; 33(27): e2008517, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34048090

RESUMEN

Worldwide, over 26 million patients suffer from heart failure (HF). One strategy aspiring to prevent or even to reverse HF is based on the transplantation of cardiac tissue-engineered (cTE) constructs. These patient-specific constructs aim to closely resemble the native myocardium and, upon implantation on the diseased tissue, support and restore cardiac function, thereby preventing the development of HF. However, cTE constructs off-the-shelf availability in the clinical arena critically depends on the development of efficient preservation methodologies. Short- and long-term preservation of cTE constructs would enable transportation and direct availability. Herein, currently available methods, from normothermic- to hypothermic- to cryopreservation, for the preservation of cardiomyocytes, whole-heart, and regenerative materials are reviewed. A theoretical foundation and recommendations for future research on developing cTE construct specific preservation methods are provided. Current research suggests that vitrification can be a promising procedure to ensure long-term cryopreservation of cTE constructs, despite the need of high doses of cytotoxic cryoprotective agents. Instead, short-term cTE construct preservation can be achieved at normothermic or hypothermic temperatures by administration of protective additives. With further tuning of these promising methods, it is anticipated that cTE construct therapy can be brought one step closer to the patient.


Asunto(s)
Criopreservación , Ingeniería de Tejidos , Animales , Corazón , Miocardio
9.
Front Cell Dev Biol ; 9: 624601, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33614654

RESUMEN

Human mesenchymal stem cells gather special interest as a universal and feasible add-on therapy for myocardial infarction (MI). In particular, human umbilical cord matrix-derived mesenchymal stromal cells (UCM-MSC) are advantageous since can be easily obtained and display high expansion potential. Using isolation protocols compliant with cell therapy, we previously showed UCM-MSC preserved cardiac function and attenuated remodeling 2 weeks after MI. In this study, UCM-MSC from two umbilical cords, UC-A and UC-B, were transplanted in a murine MI model to investigate consistency and durability of the therapeutic benefits. Both cellular products improved cardiac function and limited adverse cardiac remodeling 12 weeks post-ischemic injury, supporting sustained and long-term beneficial therapeutic effect. Donor associated variability was found in the modulation of cardiac remodeling and activation of the Akt-mTOR-GSK3ß survival pathway. In vitro, the two cell products displayed similar ability to induce the formation of vessel-like structures and comparable transcriptome in normoxia and hypoxia, apart from UCM-MSCs proliferation and expression differences in a small subset of genes associated with MHC Class I. These findings support that UCM-MSC are strong candidates to assist the treatment of MI whilst calling for the discussion on methodologies to characterize and select best performing UCM-MSC before clinical application.

10.
EMBO Rep ; 22(3): e52162, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33586846

RESUMEN

Understanding the mechanisms by which natural anti-freeze proteins protect cells and tissues from cold could help to improve the availability of donor organs for transplantation.


Asunto(s)
Proteínas Anticongelantes
11.
Methods ; 190: 55-62, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32603825

RESUMEN

Cardiovascular diseases remain the leading cause of death, largely due to the limited regenerative capacity of the adult mammalian heart. Yet, neonatal mammals were shown to regenerate the myocardium after injury by increasing the proliferation of pre-existing cardiomyocytes. Re-activation of cardiomyocyte proliferation in adulthood has been considered a promising strategy to improve cardiac response to injury. Notwithstanding, quantification of cardiomyocyte proliferation, which occurs at a very low rate, is hampered by inefficient or unreliable techniques. Herein, we propose an optimized protocol to unequivocally assess cardiomyocyte proliferation and/or cardiomyocyte number in the myocardium. Resorting to a stereological approach we estimate the number of cardiomyocytes using representative thick sections of left ventricle fragments. This protocol overcomes the need for spatial-temporal capture of cardiomyocyte proliferation events by focusing instead on the quantification of the outcome of this process. In addition, assessment of cardiomyocyte nucleation avoids overestimation of cardiomyocyte proliferation due to increased binucleation. By applying this protocol, we were able to previously show that apical resection triggers proliferation of pre-existing cardiomyocytes generating hearts with more cardiomyocytes. Likewise, the protocol will be useful for any study aiming at evaluating the impact of neomyogenic therapies.


Asunto(s)
Corazón , Miocitos Cardíacos , Animales , Proliferación Celular , Ventrículos Cardíacos , Miocardio , Regeneración
12.
Semin Cell Dev Biol ; 112: 16-26, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32591270

RESUMEN

Cardiac chamber walls contain large numbers of non-contractile interstitial cells, including fibroblasts, endothelial cells, pericytes and significant populations of blood lineage-derived cells. Blood cells first colonize heart tissues a few days before birth, although their recruitment from the bloodstream to the cardiac interstitium is continuous and extends throughout adult life. The bone marrow, as the major hematopoietic site of adult individuals, is in charge of renewing all circulating cell types, and it therefore plays a pivotal role in the incorporation of blood cells to the heart. Bone marrow-derived cells are instrumental to tissue homeostasis in the steady-state heart, and are major effectors in cardiac disease progression. This review will provide a comprehensive approach to bone marrow-derived blood cell functions in the heart, and discuss aspects related to hot topics in the cardiovascular field like cell-based heart regeneration strategies.


Asunto(s)
Médula Ósea/fisiología , Corazón/crecimiento & desarrollo , Células Madre Hematopoyéticas/fisiología , Regeneración/fisiología , Células de la Médula Ósea/fisiología , Diferenciación Celular/genética , Linaje de la Célula/genética , Linaje de la Célula/fisiología , Células Endoteliales/fisiología , Corazón/fisiopatología , Cardiopatías/genética , Cardiopatías/fisiopatología , Humanos
13.
Biochim Biophys Acta Mol Cell Res ; 1867(3): 118515, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31362011

RESUMEN

Heart failure (HF) as a result of myocardial infarction (MI) is the leading cause of death worldwide. In contrast to the adult mammalian heart, which has low regenerative capacity, newborn mammalian and zebrafish hearts can completely regenerate after injury. Cardiac regeneration is considered to be mediated by proliferation of pre-existing cardiomyocytes (CMs) mainly located in a hypoxic niche. To find new therapies to treat HF, efforts are being made to understand the molecular pathways underlying the regenerative capacity of the heart. However, the multicellularity of the heart is important during cardiac regeneration as not only CM proliferation but also the restoration of the endothelium is imperative to prevent progression to HF. It has recently come to light that signalling from non-coding RNAs (ncRNAs) and extracellular vesicles (EVs) plays a role in the healthy and the diseased heart. Multiple studies identified differentially expressed ncRNAs after MI, making them potential therapeutic targets. In this review, we highlight the molecular interactions between endothelial cells (ECs) and CMs in cardiac regeneration and when the heart loses its regenerative capacity. We specifically emphasize the role of ncRNAs and cell-cell communication via EVs during cardiac regeneration and neovascularisation.


Asunto(s)
Insuficiencia Cardíaca/terapia , Corazón/crecimiento & desarrollo , Miocitos Cardíacos/metabolismo , Regeneración/genética , Animales , Hipoxia de la Célula/genética , Proliferación Celular/genética , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Corazón/fisiopatología , Insuficiencia Cardíaca/patología , Humanos , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Miocitos Cardíacos/fisiología , Pez Cebra/genética
14.
Cell Physiol Biochem ; 52(3): 503-516, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30897318

RESUMEN

BACKGROUND/AIMS: Vascular complications contribute significantly to the extensive morbidity and mortality rates observed in people with diabetes. Despite well known that the diabetic kidney and heart exhibit imbalanced angiogenesis, the mechanisms implicated in this angiogenic paradox remain unknown. In this study, we examined the angiogenic and metabolic gene expression profile (GEP) of endothelial cells (ECs) isolated from a mouse model with type1 diabetes mellitus (T1DM). METHODS: ECs were isolated from kidneys and hearts of healthy and streptozocin (STZ)-treated mice. RNA was then extracted for molecular studies. GEP of 84 angiogenic and 84 AMP-activated Protein Kinase (AMPK)-dependent genes were examined by microarrays. Real time PCR confirmed the changes observed in significantly altered genes. Microvessel density (MVD) was analysed by immunohistochemistry, fibrosis was assessed by the Sirius red histological staining and connective tissue growth factor (CTGF) was quantified by ELISA. RESULTS: The relative percentage of ECs and MVD were increased in the kidneys of T1DM animals whereas the opposite trend was observed in the hearts of diabetic mice. Accordingly, the majority of AMPK-associated genes were upregulated in kidneys and downregulated in hearts of these animals. Angiogenic GEP revealed significant differences in Tgfß, Notch signaling and Timp2 in both diabetic organs. These findings were in agreement with the angiogenesis histological assays. Fibrosis was augmented in both organs in diabetic as compared to healthy animals. CONCLUSION: Altogether, our findings indicate, for the first time, that T1DM heart and kidney ECs present opposite metabolic cues, which are accompanied by distinct angiogenic patterns. These findings enable the development of innovative organ-specific therapeutic strategies targeting diabetic-associated vascular disorders.


Asunto(s)
Diabetes Mellitus Experimental/patología , Células Endoteliales/metabolismo , Microvasos/fisiología , Animales , Factor de Crecimiento del Tejido Conjuntivo/análisis , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/citología , Fibrosis , Ventrículos Cardíacos/metabolismo , Riñón/citología , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microvasos/patología , Miocardio/citología , Miocardio/metabolismo , Neovascularización Patológica , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptores Notch/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/genética , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Transcriptoma , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
15.
Mol Ther ; 27(3): 584-599, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30559069

RESUMEN

Heart failure is preceded by ventricular remodeling, changes in left ventricular mass, and myocardial volume after alterations in loading conditions. Concentric hypertrophy arises after pressure overload, involves wall thickening, and forms a substrate for diastolic dysfunction. Eccentric hypertrophy develops in volume overload conditions and leads wall thinning, chamber dilation, and reduced ejection fraction. The molecular events underlying these distinct forms of cardiac remodeling are poorly understood. Here, we demonstrate that miR-148a expression changes dynamically in distinct subtypes of heart failure: while it is elevated in concentric hypertrophy, it decreased in dilated cardiomyopathy. In line, antagomir-mediated silencing of miR-148a caused wall thinning, chamber dilation, increased left ventricle volume, and reduced ejection fraction. Additionally, adeno-associated viral delivery of miR-148a protected the mouse heart from pressure-overload-induced systolic dysfunction by preventing the transition of concentric hypertrophic remodeling toward dilation. Mechanistically, miR-148a targets the cytokine co-receptor glycoprotein 130 (gp130) and connects cardiomyocyte responsiveness to extracellular cytokines by modulating the Stat3 signaling. These findings show the ability of miR-148a to prevent the transition of pressure-overload induced concentric hypertrophic remodeling toward eccentric hypertrophy and dilated cardiomyopathy and provide evidence for the existence of separate molecular programs inducing distinct forms of myocardial remodeling.


Asunto(s)
Cardiomiopatías/metabolismo , Insuficiencia Cardíaca/metabolismo , Trasplante de Corazón/métodos , MicroARNs/metabolismo , Miocardio/metabolismo , Animales , Cardiomiopatías/genética , Proliferación Celular/fisiología , Insuficiencia Cardíaca/genética , Humanos , Ratones , MicroARNs/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/fisiología , Remodelación Ventricular/genética , Remodelación Ventricular/fisiología
16.
Stem Cell Reports ; 10(3): 860-874, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29503089

RESUMEN

So far, opposing outcomes have been reported following neonatal apex resection in mice, questioning the validity of this injury model to investigate regenerative mechanisms. We performed a systematic evaluation, up to 180 days after surgery, of the pathophysiological events activated upon apex resection. In response to cardiac injury, we observed increased cardiomyocyte proliferation in remote and apex regions, neovascularization, and local fibrosis. In adulthood, resected hearts remain consistently shorter and display permanent fibrotic tissue deposition in the center of the resection plane, indicating limited apex regrowth. However, thickening of the left ventricle wall, explained by an upsurge in cardiomyocyte proliferation during the initial response to injury, compensated cardiomyocyte loss and supported normal systolic function. Thus, apex resection triggers both regenerative and reparative mechanisms, endorsing this injury model for studies aimed at promoting cardiomyocyte proliferation and/or downplaying fibrosis.


Asunto(s)
Proliferación Celular/fisiología , Fibrosis/fisiopatología , Corazón/fisiología , Miocitos Cardíacos/fisiología , Neovascularización Patológica/fisiopatología , Recuperación de la Función/fisiología , Animales , Animales Recién Nacidos , Lesiones Cardíacas/fisiopatología , Ventrículos Cardíacos/fisiopatología , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , Regeneración/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...