Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Regen ; 13(1): 10, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38649624

RESUMEN

Human cardiac and other organoids have recently emerged as a groundbreaking tool for advancing our understanding the developmental biology of human organs. A recent paper from Sasha Mendjan's laboratory published in the journal Cell on December 7, 2023, reported the generation of multi-chamber cardioids from human pluripotent stem cells, a transformative technology in the field of cardiology. In this short highlight paper, we summarize their findings. Their cardioids remarkably recapitulate the complexity of the human embryonic heart, including tissue architecture, cellular diversity, and functionality providing an excellent in vitro model for investigation of human heart development, disease modeling, precision medicine, and regenerative medicine. Thus, generating cardioids is an important step forward for understanding human heart development and developing potential therapies for heart diseases.

2.
Cell Regen ; 12(1): 36, 2023 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-37938438

RESUMEN

Whole-body regeneration is a multifaceted process that reinstates a body to its initial three-dimension size and structure after resection injury. It is well-known that signaling waves such as calcium and extracellular signal-related kinase (ERK) signaling waves can efficiently transmit information between tissues or cells. However, the mechanisms responsible for coordinating wound responses over long distances are largely unexplored. A recent study has reported that the propagation of ERK signaling waves via longitudinal body-wall muscles play an essential role in wound response and whole-body regeneration in planarians, underscoring the significance of feedback interactions between spatially distinct tissues during whole-body regeneration over long distances. These findings not only address the central questions of regenerative biology but also have potential implications for regenerative medicine.

3.
Cell Regen ; 12(1): 25, 2023 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-37466803

RESUMEN

Ischemia-reperfusion injury occurs after reperfusion treatment for patients suffering myocardial infarction, however the underlying mechanisms are incompletely understood and effective pharmacological interventions are limited. Here, we report the identification and characterization of the FDA-approved drug disulfiram (DSF) as a cardioprotective compound. By applying high-throughput chemical screening, we found that DSF decreased H2O2-induced cardiomyocyte death by inhibiting Gasdermin D, but not ALDH1, in cardiomyocytes. Oral gavage of DSF decreased myocardial infarct size and improved heart function after myocardial ischemia-reperfusion injury in rats. Therefore, this work reveals DSF as a potential therapeutic compound for the treatment of ischemic heart disease.

5.
Protein Cell ; 14(5): 350-368, 2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-37155312

RESUMEN

Mammals exhibit limited heart regeneration ability, which can lead to heart failure after myocardial infarction. In contrast, zebrafish exhibit remarkable cardiac regeneration capacity. Several cell types and signaling pathways have been reported to participate in this process. However, a comprehensive analysis of how different cells and signals interact and coordinate to regulate cardiac regeneration is unavailable. We collected major cardiac cell types from zebrafish and performed high-precision single-cell transcriptome analyses during both development and post-injury regeneration. We revealed the cellular heterogeneity as well as the molecular progress of cardiomyocytes during these processes, and identified a subtype of atrial cardiomyocyte exhibiting a stem-like state which may transdifferentiate into ventricular cardiomyocytes during regeneration. Furthermore, we identified a regeneration-induced cell (RIC) population in the epicardium-derived cells (EPDC), and demonstrated Angiopoietin 4 (Angpt4) as a specific regulator of heart regeneration. angpt4 expression is specifically and transiently activated in RIC, which initiates a signaling cascade from EPDC to endocardium through the Tie2-MAPK pathway, and further induces activation of cathepsin K in cardiomyocytes through RA signaling. Loss of angpt4 leads to defects in scar tissue resolution and cardiomyocyte proliferation, while overexpression of angpt4 accelerates regeneration. Furthermore, we found that ANGPT4 could enhance proliferation of neonatal rat cardiomyocytes, and promote cardiac repair in mice after myocardial infarction, indicating that the function of Angpt4 is conserved in mammals. Our study provides a mechanistic understanding of heart regeneration at single-cell precision, identifies Angpt4 as a key regulator of cardiomyocyte proliferation and regeneration, and offers a novel therapeutic target for improved recovery after human heart injuries.


Asunto(s)
Infarto del Miocardio , Pez Cebra , Humanos , Ratones , Ratas , Proliferación Celular , Corazón/fisiología , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Pericardio/metabolismo , Análisis de la Célula Individual , Pez Cebra/metabolismo , Animales
6.
NPJ Regen Med ; 8(1): 21, 2023 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-37029137

RESUMEN

Myocardial Brg1 is essential for heart regeneration in zebrafish, but it remains unknown whether and how endothelial Brg1 plays a role in heart regeneration. Here, we found that both brg1 mRNA and protein were induced in cardiac endothelial cells after ventricular resection and endothelium-specific overexpression of dominant-negative Xenopus Brg1 (dn-xbrg1) inhibited myocardial proliferation and heart regeneration and increased cardiac fibrosis. RNA-seq and ChIP-seq analysis revealed that endothelium-specific overexpression of dn-xbrg1 changed the levels of H3K4me3 modifications in the promoter regions of the zebrafish genome and induced abnormal activation of Notch family genes upon injury. Mechanistically, Brg1 interacted with lysine demethylase 7aa (Kdm7aa) to fine-tune the level of H3K4me3 within the promoter regions of Notch family genes and thus regulated notch gene transcription. Together, this work demonstrates that the Brg1-Kdm7aa-Notch axis in cardiac endothelial cells, including the endocardium, regulates myocardial proliferation and regeneration via modulating the H3K4me3 of the notch promoters in zebrafish.

7.
Dis Model Mech ; 16(5)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36478044

RESUMEN

Acute myocardial infarction (MI) results in loss of cardiomyocytes and abnormal cardiac remodeling with severe inflammation and fibrosis. However, how cardiac repair can be achieved by timely resolution of inflammation and cardiac fibrosis remains incompletely understood. Our previous findings have shown that dual-specificity phosphatase 6 (DUSP6) is a regeneration repressor from zebrafish to rats. In this study, we found that intravenous administration of the DUSP6 inhibitor (E)-2-benzylidene-3-(cyclohexylamino)-2,3-dihydro-1H-inden-1-one (BCI) improved heart function and reduced cardiac fibrosis in MI rats. Mechanistic analysis revealed that BCI attenuated macrophage inflammation through NF-κB and p38 signaling, independent of DUSP6 inhibition, leading to the downregulation of various cytokines and chemokines. In addition, BCI suppressed differentiation-related signaling pathways and decreased bone-marrow cell differentiation into macrophages through inhibiting DUSP6. Furthermore, intramyocardial injection of poly (D, L-lactic-co-glycolic acid)-loaded BCI after MI had a notable effect on cardiac repair. In summary, BCI improves heart function and reduces abnormal cardiac remodeling by inhibiting macrophage formation and inflammation post-MI, thus providing a promising pro-drug candidate for the treatment of MI and related heart diseases. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Infarto del Miocardio , Animales , Ratas , Fosfatasa 6 de Especificidad Dual , Fibrosis , Infarto del Miocardio/complicaciones , Infarto del Miocardio/tratamiento farmacológico , Remodelación Ventricular
8.
Nat Commun ; 13(1): 6672, 2022 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-36335128

RESUMEN

Dual-specificity phosphatase 6 (DUSP6) serves a specific and conserved function on the dephosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2). We previously identified Dusp6 as a regenerative repressor during zebrafish heart regeneration, therefore we propose to investigate the role of this repressor in mammalian cardiac repair. Utilizing a rat strain harboring Dusp6 nonsense mutation, rat neutrophil-cardiomyocyte co-culture, bone marrow transplanted rats and neutrophil-specific Dusp6 knockout mice, we find that Dusp6 deficiency improves cardiac outcomes by predominantly attenuating neutrophil-mediated myocardial damage in acute inflammatory phase after myocardial infarction. Mechanistically, Dusp6 is transcriptionally activated by p38-C/EBPß signaling and acts as an effector for maintaining p-p38 activity by down-regulating pERK and p38-targeting phosphatases DUSP1/DUSP16. Our findings provide robust animal models and novel insights for neutrophil-mediated cardiac damage and demonstrate the potential of DUSP6 as a therapeutic target for post-MI cardiac remodeling and other relevant inflammatory diseases.


Asunto(s)
Infarto del Miocardio , Animales , Ratones , Ratas , Fosfatasa 6 de Especificidad Dual , Ratones Noqueados , Infarto del Miocardio/genética , Miocardio , Miocitos Cardíacos , Neutrófilos
9.
Sci Rep ; 12(1): 9810, 2022 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-35697754

RESUMEN

Rheumatoid arthritis (RA) is chronic systemic disease that can cause joint damage, disability and destructive polyarthritis. Current diagnosis of RA is based on a combination of clinical and laboratory features. However, RA diagnosis can be difficult at its disease onset on account of overlapping symptoms with other arthritis, so early recognition and diagnosis of RA permit the better management of patients. In order to improve the medical diagnosis of RA and evaluate the effects of different clinical features on RA diagnosis, we applied an artificial neural network (ANN) as the training algorithm, and used fivefold cross-validation to evaluate its performance. From each sample, we obtained data on 6 features: age, sex, rheumatoid factor, anti-citrullinated peptide antibody (CCP), 14-3-3η, and anti-carbamylated protein (CarP) antibodies. After training, this ANN model assigned each sample a probability for being either an RA patient or a non-RA patient. On the validation dataset, the F1 for all samples by this ANN model was 0.916, which was higher than the 0.906 we previously reported using an optimal threshold algorithm. Therefore, this ANN algorithm not only improved the accuracy of RA diagnosis, but also revealed that anti-CCP had the greatest effect while age and anti-CarP had a weaker on RA diagnosis.


Asunto(s)
Artritis Reumatoide , Autoanticuerpos , Artritis Reumatoide/diagnóstico , Humanos , Redes Neurales de la Computación , Péptidos Cíclicos , Factor Reumatoide
10.
Cell Stem Cell ; 29(4): 545-558.e13, 2022 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-35395187

RESUMEN

Zebrafish and mammalian neonates possess robust cardiac regeneration via the induction of endogenous cardiomyocyte (CM) proliferation, but adult mammalian hearts have very limited regenerative potential. Developing small molecules for inducing adult mammalian heart regeneration has had limited success. We report a chemical cocktail of five small molecules (5SM) that promote adult CM proliferation and heart regeneration. A high-content chemical screen, along with an algorithm-aided prediction of small-molecule interactions, identified 5SM that efficiently induced CM cell cycle re-entry and cytokinesis. Intraperitoneal delivery of 5SM reversed the loss of heart function, induced CM proliferation, and decreased cardiac fibrosis after rat myocardial infarction. Mechanistically, 5SM potentially targets α1 adrenergic receptor, JAK1, DYRKs, PTEN, and MCT1 and is connected to lactate-LacRS2 signaling, leading to CM metabolic switching toward glycolysis/biosynthesis and CM de-differentiation before entering the cell-cycle. Our work sheds lights on the understanding CM regenerative mechanisms and opens therapeutic avenues for repairing the heart.


Asunto(s)
Infarto del Miocardio , Miocitos Cardíacos , Animales , Proliferación Celular , Corazón , Mamíferos , Infarto del Miocardio/tratamiento farmacológico , Miocitos Cardíacos/metabolismo , Ratas , Transducción de Señal , Pez Cebra
11.
STAR Protoc ; 3(4): 101903, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36595956

RESUMEN

Discovery of small molecules promoting cardiomyocyte proliferation is important for heart regeneration and related heart disease. Here, we describe a protocol to isolate neonatal rat and mouse cardiomyocytes, infect cardiomyocytes with Tnnt2-mAG-hGeminin (1/110) or Tnnt2-Cre adenovirus, and identify small molecules that promote cardiomyocyte proliferation by high-content microscopy. This protocol can be modified to investigate other pro-proliferation factors in cardiomyocytes and other cell types. For complete details on the use and execution of this protocol, please refer to Du et al. (2022).1.


Asunto(s)
Miocitos Cardíacos , Troponina T , Animales , Ratones , Ratas , Miocitos Cardíacos/metabolismo , Proliferación Celular
12.
Cell Regen ; 10(1): 34, 2021 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-34725734

RESUMEN

Organ regeneration is an important, fascinating, and old topic while much remains unknown in spite of extensive investigations for decades. From March 25th to 27th, 2021, the Third Chinese Symposium on Organ Regeneration took place in the beautiful ocean city of Zhoushan, Zhejiang, China. This biennial conference attracted ~ 300 academic attendees: students, postdoctoral fellows, and principal investigators, in addition to few industrial investigators. The mixed live and virtual talks covered the broad field of organ regeneration from different animal organisms to human organoids, and concluded with some impressive advances on inflammatory signaling, regenerative signaling mechanisms, new technologies, and applications for organ regeneration.

13.
J Cell Sci ; 134(21)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34622921

RESUMEN

Cardiac looping and trabeculation are key processes during cardiac chamber maturation. However, the underlying mechanisms remain incompletely understood. Here, we report the isolation, cloning and characterization of the proprotein convertase furina from the cardiovascular mutant loft in zebrafish. loft is an ethylnitrosourea-induced mutant and has evident defects in the cardiac outflow tract, heart looping and trabeculation, the craniofacial region and pharyngeal arch arteries. Positional cloning revealed that furina mRNA was barely detectable in loft mutants, and loft failed to complement the TALEN-induced furina mutant pku338, confirming that furina is responsible for the loft mutant phenotypes. Mechanistic studies demonstrated that Notch reporter Tg(tp1:mCherry) signals were largely eliminated in mutant hearts, and overexpression of the Notch intracellular domain partially rescued the mutant phenotypes, probably due to the lack of Furina-mediated cleavage processing of Notch1b proteins, the only Notch receptor expressed in the heart. Together, our data suggest a potential post-translational modification of Notch1b proteins via the proprotein convertase Furina in the heart, and unveil the function of the Furina-Notch1b axis in cardiac looping and trabeculation in zebrafish, and possibly in other organisms.


Asunto(s)
Proproteína Convertasas , Proteínas de Pez Cebra , Pez Cebra , Animales , Corazón , Organogénesis/genética , Receptores Notch/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
14.
Nanoscale ; 13(35): 14636-14643, 2021 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-34558568

RESUMEN

Mimicking and leveraging biological structures and materials provide important approaches to develop functional vehicles for drug delivery. Taking advantage of the affinity and adhesion between the activated endothelial cells and innate immune cells during inflammatory responses, hybrid polyester nanoparticles coated with endothelial cell membranes (EM-P) containing adhesion molecules were fabricated and their capability as vehicles to travel to the acute injury sites through leukocyte-mediated processes was investigated. The in vivo studies and quantitative analyses performed through the lung-inflammation mouse models demonstrated that the EM-Ps preferentially interacted with the neutrophils and monocytes in the circulation and the cellular membrane-based biosurface improved the nanoparticle transportation to the inflamed lung possibly via the motility of neutrophils. Utilizing the transgenic zebrafish model, the leukocyte-mediated transportation and biodistribution of EM-Ps were further visualized in real time at the whole-organism level. Endothelial membranes provided a new biosurface for developing biomimetic vehicles to allow the immune cell-mediated transportation and may enable advanced systems for active and highly efficient drug delivery.


Asunto(s)
Células Endoteliales , Nanopartículas , Animales , Leucocitos , Ratones , Distribución Tisular , Pez Cebra
15.
Int J Biol Sci ; 17(9): 2157-2166, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34239346

RESUMEN

Adult mammalian hearts show limited capacity to proliferate after injury, while zebrafish are capable to completely regenerate injured hearts through the proliferation of spared cardiomyocytes. BMP and Notch signaling pathways have been implicated in cardiomyocyte proliferation during zebrafish heart regeneration. However, the molecular mechanism underneath this process as well as the interaction between these two pathways remains to be further explored. In this study we showed BMP signaling was activated after ventricle ablation and acted epistatic downstream of Notch signaling. Inhibition of both signaling pathways differentially influenced ventricle regeneration and cardiomyocyte proliferation, as revealed by time-lapse analysis using a cardiomyocyte-specific FUCCI (fluorescent ubiquitylation-based cell cycle indicator) system. Further experiments revealed that inhibition of BMP and Notch signaling led to cell-cycle arrest at different phases. Overall, our results shed light on the interaction between BMP and Notch signaling pathways and their functions in cardiomyocyte proliferation during cardiac regeneration.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Corazón/fisiología , Miocitos Cardíacos/citología , Receptores Notch/metabolismo , Regeneración , Transducción de Señal , Animales , Puntos de Control del Ciclo Celular , Proliferación Celular , Miocitos Cardíacos/metabolismo , Ubiquitinación , Pez Cebra/fisiología
17.
Front Cell Dev Biol ; 9: 632372, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33816481

RESUMEN

Unlike mammals, zebrafish can regenerate injured hearts even in the adult stage. Cardiac regeneration requires the coordination of cardiomyocyte (CM) proliferation and migration. The TGF-ß/Smad3 signaling pathway has been implicated in cardiac regeneration, but the molecular mechanisms by which this pathway regulates CM proliferation and migration have not been fully illustrated. Here, we investigated the function of TGF-ß/Smad3 signaling in a zebrafish model of ventricular ablation. Multiple components of this pathway were upregulated/activated after injury. Utilizing a specific inhibitor of Smad3, we detected an increased ratio of unrecovered hearts. Transcriptomic analysis suggested that the TGF-ß/Smad3 signaling pathway could affect CM proliferation and migration. Further analysis demonstrated that the CM cell cycle was disrupted and the epithelial-mesenchymal transition (EMT)-like response was impaired, which limited cardiac regeneration. Altogether, our study reveals an important function of TGF-ß/Smad3 signaling in CM cell cycle progression and EMT process during zebrafish ventricle regeneration.

18.
Cell Regen ; 10(1): 13, 2021 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-33821373

RESUMEN

Heart regeneration is a fascinating and complex biological process. Decades of intensive studies have revealed a sophisticated molecular network regulating cardiac regeneration in the zebrafish and neonatal mouse heart. Here, we review both the classical and recent literature on the molecular and cellular mechanisms underlying heart regeneration, with a particular focus on how injury triggers the cell-cycle re-entry of quiescent cardiomyocytes to replenish their massive loss after myocardial infarction or ventricular resection. We highlight several important signaling pathways for cardiomyocyte proliferation and propose a working model of how these injury-induced signals promote cardiomyocyte proliferation. Thus, this concise review provides up-to-date research progresses on heart regeneration for investigators in the field of regeneration biology.

19.
Cell Regen ; 9(1): 23, 2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-33258016

RESUMEN

Some lower vertebrates such as zebrafish and axolotl have incredible cardiac regenerative potential while mammals have very limited ones. Comparative studies among species have revealed that cardiomyocyte polyploidy, endothermy, and injury-induced activation of certain transcriptional factors including AP1 complexes are critical for cardiomyocyte proliferation and heart regeneration during animal evolution. Gaining insights into these evolutionarily conserved mechanisms will likely lead to achieving heart regeneration in non-regenerative mammals including humans.

20.
Cell Discov ; 6: 74, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33133634

RESUMEN

It remains challenging to construct a complete cell lineage map of the origin of vascular endothelial cells in any vertebrate embryo. Here, we report the application of in toto light-sheet fluorescence imaging of embryos to trace the origin of vascular endothelial cells (ECs) at single-cell resolution in zebrafish. We first adapted a previously reported method to embryo mounting and light-sheet imaging, created an alignment, fusion, and extraction all-in-one software (AFEIO) for processing big data, and performed quantitative analysis of cell lineage relationships using commercially available Imaris software. Our data revealed that vascular ECs originated from broad regions of the gastrula along the dorsal-ventral and anterior-posterior axes, of which the dorsal-anterior cells contributed to cerebral ECs, the dorsal-lateral cells to anterior trunk ECs, and the ventral-lateral cells to posterior trunk and tail ECs. Therefore, this work, to our knowledge, charts the first comprehensive map of the gastrula origin of vascular ECs in zebrafish, and has potential applications for studying the origin of any embryonic organs in zebrafish and other model organisms.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA