Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(28): e2200342119, 2022 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-35867745

RESUMEN

Teleost fishes and urodele amphibians can regenerate amputated appendages, whereas this ability is restricted to digit tips in adult mammals. One key component of appendage regeneration is reinnervation of the wound area. However, how innervation is regulated in injured appendages of adult vertebrates has seen limited research attention. From a forward genetics screen for temperature-sensitive defects in zebrafish fin regeneration, we identified a mutation that disrupted regeneration while also inducing paralysis at the restrictive temperature. Genetic mapping and complementation tests identify a mutation in the major neuronal voltage-gated sodium channel (VGSC) gene scn8ab. Conditional disruption of scn8ab impairs early regenerative events, including blastema formation, but does not affect morphogenesis of established regenerates. Whereas scn8ab mutations reduced neural activity as expected, they also disrupted axon regrowth and patterning in fin regenerates, resulting in hypoinnervation. Our findings indicate that the activity of VGSCs plays a proregenerative role by promoting innervation of appendage stumps.


Asunto(s)
Aletas de Animales , Canal de Sodio Activado por Voltaje NAV1.6 , Regeneración , Proteínas de Pez Cebra , Pez Cebra , Aletas de Animales/inervación , Aletas de Animales/fisiología , Animales , Mutación , Canal de Sodio Activado por Voltaje NAV1.6/genética , Canal de Sodio Activado por Voltaje NAV1.6/fisiología , Regeneración/genética , Regeneración/fisiología , Pez Cebra/genética , Pez Cebra/fisiología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
2.
Development ; 148(2)2021 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-33234717

RESUMEN

The ability of zebrafish to heal their heart after injury makes them an attractive model for investigating the mechanisms governing the regenerative process. In this study, we show that the gene cellular communication network factor 2a (ccn2a), previously known as ctgfa, is induced in endocardial cells in the injured tissue and regulates CM proliferation and repopulation of the damaged tissue. We find that, whereas in wild-type animals, CMs track along the newly formed blood vessels that revascularize the injured tissue, in ccn2a mutants CM proliferation and repopulation are disrupted, despite apparently unaffected revascularization. In addition, we find that ccn2a overexpression enhances CM proliferation and improves the resolution of transient collagen deposition. Through loss- and gain-of-function as well as pharmacological approaches, we provide evidence that Ccn2a is necessary for and promotes heart regeneration by enhancing the expression of pro-regenerative extracellular matrix genes, and by inhibiting the chemokine receptor gene cxcr3.1 through a mechanism involving Tgfß/pSmad3 signaling. Thus, Ccn2a positively modulates the innate regenerative response of the adult zebrafish heart.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Corazón/fisiopatología , Regeneración , Proteínas de Pez Cebra/metabolismo , Pez Cebra/fisiología , Animales , Núcleo Celular/metabolismo , Proliferación Celular , Factor de Crecimiento del Tejido Conjuntivo/genética , Vasos Coronarios/metabolismo , Endocardio/patología , Endocardio/fisiopatología , Matriz Extracelular/genética , Regulación del Desarrollo de la Expresión Génica , Mutación/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fosforilación , Transporte de Proteínas , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas de Pez Cebra/genética
3.
Nature ; 532(7598): 201-6, 2016 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-27049946

RESUMEN

How tissue regeneration programs are triggered by injury has received limited research attention. Here we investigate the existence of enhancer regulatory elements that are activated in regenerating tissue. Transcriptomic analyses reveal that leptin b (lepb) is highly induced in regenerating hearts and fins of zebrafish. Epigenetic profiling identified a short DNA sequence element upstream and distal to lepb that acquires open chromatin marks during regeneration and enables injury-dependent expression from minimal promoters. This element could activate expression in injured neonatal mouse tissues and was divisible into tissue-specific modules sufficient for expression in regenerating zebrafish fins or hearts. Simple enhancer-effector transgenes employing lepb-linked sequences upstream of pro- or anti-regenerative factors controlled the efficacy of regeneration in zebrafish. Our findings provide evidence for 'tissue regeneration enhancer elements' (TREEs) that trigger gene expression in injury sites and can be engineered to modulate the regenerative potential of vertebrate organs.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Especificidad de Órganos/genética , Regeneración/genética , Regeneración/fisiología , Cicatrización de Heridas/genética , Pez Cebra/genética , Pez Cebra/fisiología , Acetilación , Aletas de Animales/lesiones , Aletas de Animales/metabolismo , Animales , Animales Recién Nacidos , Proliferación Celular , Ensamble y Desensamble de Cromatina/genética , Epigénesis Genética/genética , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Corazón , Histonas/química , Histonas/metabolismo , Leptina/biosíntesis , Leptina/genética , Lisina/metabolismo , Masculino , Ratones , Miocitos Cardíacos/citología , Regiones Promotoras Genéticas/genética , Transgenes/genética , Proteínas de Pez Cebra/genética
4.
Nature ; 522(7555): 226-230, 2015 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-25938716

RESUMEN

In response to cardiac damage, a mesothelial tissue layer enveloping the heart called the epicardium is activated to proliferate and accumulate at the injury site. Recent studies have implicated the epicardium in multiple aspects of cardiac repair: as a source of paracrine signals for cardiomyocyte survival or proliferation; a supply of perivascular cells and possibly other cell types such as cardiomyocytes; and as a mediator of inflammation. However, the biology and dynamism of the adult epicardium is poorly understood. To investigate this, we created a transgenic line to ablate the epicardial cell population in adult zebrafish. Here we find that genetic depletion of the epicardium after myocardial loss inhibits cardiomyocyte proliferation and delays muscle regeneration. The epicardium vigorously regenerates after its ablation, through proliferation and migration of spared epicardial cells as a sheet to cover the exposed ventricular surface in a wave from the chamber base towards its apex. By reconstituting epicardial regeneration ex vivo, we show that extirpation of the bulbous arteriosus-a distinct, smooth-muscle-rich tissue structure that distributes outflow from the ventricle-prevents epicardial regeneration. Conversely, experimental repositioning of the bulbous arteriosus by tissue recombination initiates epicardial regeneration and can govern its direction. Hedgehog (Hh) ligand is expressed in the bulbous arteriosus, and treatment with a Hh signalling antagonist arrests epicardial regeneration and blunts the epicardial response to muscle injury. Transplantation of Sonic hedgehog (Shh)-soaked beads at the ventricular base stimulates epicardial regeneration after bulbous arteriosus removal, indicating that Hh signalling can substitute for the influence of the outflow tract. Thus, the ventricular epicardium has pronounced regenerative capacity, regulated by the neighbouring cardiac outflow tract and Hh signalling. These findings extend our understanding of tissue interactions during regeneration and have implications for mobilizing epicardial cells for therapeutic heart repair.


Asunto(s)
Lesiones Cardíacas/metabolismo , Proteínas Hedgehog/metabolismo , Pericardio/fisiología , Regeneración/fisiología , Transducción de Señal , Proteínas de Pez Cebra/metabolismo , Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente , Proliferación Celular/genética , Femenino , Proteínas Hedgehog/genética , Masculino , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Pericardio/citología , Regeneración/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
5.
Development ; 143(2): 232-43, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26657776

RESUMEN

In contrast to mammals, adult zebrafish have a high capacity to regenerate damaged or lost myocardium through proliferation of cardiomyocytes spared from damage. The epicardial sheet covering the heart is activated by injury and aids muscle regeneration through paracrine effects and as a multipotent cell source, and has received recent attention as a target in cardiac repair strategies. Although it is recognized that epicardium is required for muscle regeneration and itself has high regenerative potential, the extent of cellular heterogeneity within epicardial tissue is largely unexplored. Here, we performed transcriptome analysis on dozens of epicardial lineage cells purified from zebrafish harboring a transgenic reporter for the pan-epicardial gene tcf21. Hierarchical clustering analysis suggested the presence of at least three epicardial cell subsets defined by expression signatures. We validated many new pan-epicardial and epicardial markers by alternative expression assays. Additionally, we explored the function of the scaffolding protein and main component of caveolae, caveolin 1 (cav1), which was present in each epicardial subset. In BAC transgenic zebrafish, cav1 regulatory sequences drove strong expression in ostensibly all epicardial cells and in coronary vascular endothelial cells. Moreover, cav1 mutant zebrafish generated by genome editing showed grossly normal heart development and adult cardiac anatomy, but displayed profound defects in injury-induced cardiomyocyte proliferation and heart regeneration. Our study defines a new platform for the discovery of epicardial lineage markers, genetic tools, and mechanisms of heart regeneration.


Asunto(s)
Caveolina 1/metabolismo , Corazón/fisiología , Pericardio/citología , Regeneración/fisiología , Animales , Caveolina 1/genética , Miocitos Cardíacos/citología , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
6.
Dev Biol ; 382(2): 427-35, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23988577

RESUMEN

Unlike adult mammals, adult zebrafish vigorously regenerate lost heart muscle in response to injury. The epicardium, a mesothelial cell layer enveloping the myocardium, is activated to proliferate after cardiac injury and can contribute vascular support cells or provide mitogens to regenerating muscle. Here, we applied proteomics to identify secreted proteins that are associated with heart regeneration. We found that Fibronectin, a main component of the extracellular matrix, is induced and deposited after cardiac damage. In situ hybridization and transgenic reporter analyses indicated that expression of two fibronectin paralogues, fn1 and fn1b, are induced by injury in epicardial cells, while the itgb3 receptor is induced in cardiomyocytes near the injury site. fn1, the more dynamic of these paralogs, is induced chamber-wide within one day of injury before localizing epicardial Fn1 synthesis to the injury site. fn1 loss-of-function mutations disrupted zebrafish heart regeneration, as did induced expression of a dominant-negative Fibronectin cassette, defects that were not attributable to direct inhibition of cardiomyocyte proliferation. These findings reveal a new role for the epicardium in establishing an extracellular environment that supports heart regeneration.


Asunto(s)
Fibronectinas/metabolismo , Pericardio/fisiología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente , Proliferación Celular , Fibronectinas/genética , Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Regeneración , Proteínas de Pez Cebra/genética
7.
Development ; 138(16): 3421-30, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21752928

RESUMEN

Natural models of heart regeneration in lower vertebrates such as zebrafish are based on invasive surgeries causing mechanical injuries that are limited in size. Here, we created a genetic cell ablation model in zebrafish that facilitates inducible destruction of a high percentage of cardiomyocytes. Cell-specific depletion of over 60% of the ventricular myocardium triggered signs of cardiac failure that were not observed after partial ventricular resection, including reduced animal exercise tolerance and sudden death in the setting of stressors. Massive myocardial loss activated robust cellular and molecular responses by endocardial, immune, epicardial and vascular cells. Destroyed cardiomyocytes fully regenerated within several days, restoring cardiac anatomy, physiology and performance. Regenerated muscle originated from spared cardiomyocytes that acquired ultrastructural and electrophysiological characteristics of de-differentiation and underwent vigorous proliferation. Our study indicates that genetic depletion of cardiomyocytes, even at levels so extreme as to elicit signs of cardiac failure, can be reversed by natural regenerative capacity in lower vertebrates such as zebrafish.


Asunto(s)
Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Corazón/fisiología , Miocitos Cardíacos/citología , Regeneración , Pez Cebra/genética , Pez Cebra/fisiología , Animales , Muerte Celular
8.
Dev Cell ; 48(6): 853-863.e5, 2019 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-30713073

RESUMEN

Attaining proper organ size during development and regeneration hinges on the activity of mitogenic factors. Here, we performed a large-scale chemical screen in embryonic zebrafish to identify cardiomyocyte mitogens. Although commonly considered anti-proliferative, vitamin D analogs like alfacalcidol had rapid, potent mitogenic effects on embryonic and adult cardiomyocytes in vivo. Moreover, pharmacologic or genetic manipulation of vitamin D signaling controlled proliferation in multiple adult cell types and dictated growth rates in embryonic and juvenile zebrafish. Tissue-specific modulation of vitamin D receptor (VDR) signaling had organ-restricted effects, with cardiac VDR activation causing cardiomegaly. Alfacalcidol enhanced the regenerative response of injured zebrafish hearts, whereas VDR blockade inhibited regeneration. Alfacalcidol activated cardiac expression of genes associated with ErbB2 signaling, while ErbB2 inhibition blunted its effects on cell proliferation. Our findings identify vitamin D as mitogenic for cardiomyocytes and other cell types in zebrafish and indicate a mechanism to regulate organ size and regeneration.


Asunto(s)
Corazón/anatomía & histología , Corazón/fisiología , Miocitos Cardíacos/citología , Regeneración/efectos de los fármacos , Vitamina D/farmacología , Pez Cebra/anatomía & histología , Pez Cebra/fisiología , Animales , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Embrión no Mamífero/citología , Embrión no Mamífero/efectos de los fármacos , Corazón/efectos de los fármacos , Mitógenos/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Tamaño de los Órganos/efectos de los fármacos , Especificidad de Órganos , Transducción de Señal/efectos de los fármacos , Pez Cebra/embriología , Proteínas de Pez Cebra/metabolismo
9.
Cell Rep ; 22(8): 2026-2038, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29466731

RESUMEN

The spine is a segmented axial structure made of alternating vertebral bodies (centra) and intervertebral discs (IVDs) assembled around the notochord. Here, we show that, prior to centra formation, the outer epithelial cell layer of the zebrafish notochord, the sheath, segments into alternating domains corresponding to the prospective centra and IVD areas. This process occurs sequentially in an anteroposterior direction via the activation of Notch signaling in alternating segments of the sheath, which transition from cartilaginous to mineralizing domains. Subsequently, osteoblasts are recruited to the mineralized domains of the notochord sheath to form mature centra. Tissue-specific manipulation of Notch signaling in sheath cells produces notochord segmentation defects that are mirrored in the spine. Together, our findings demonstrate that notochord sheath segmentation provides a template for vertebral patterning in the zebrafish spine.


Asunto(s)
Tipificación del Cuerpo , Notocorda/embriología , Columna Vertebral/embriología , Pez Cebra/embriología , Animales , Cartílago/metabolismo , Regulación del Desarrollo de la Expresión Génica , Morfogénesis , Osteoblastos/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Somitos/metabolismo
10.
Dev Cell ; 40(4): 392-404.e5, 2017 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-28245924

RESUMEN

Chromatin regulation is a principal mechanism governing animal development, yet it is unclear to what extent structural changes in chromatin underlie tissue regeneration. Non-mammalian vertebrates such as zebrafish activate cardiomyocyte (CM) division after tissue damage to regenerate lost heart muscle. Here, we generated transgenic zebrafish expressing a biotinylatable H3.3 histone variant in CMs and derived cell-type-specific profiles of histone replacement. We identified an emerging program of putative enhancers that revise H3.3 occupancy during regeneration, overlaid upon a genome-wide reduction of H3.3 from promoters. In transgenic reporter lines, H3.3-enriched elements directed gene expression in subpopulations of CMs. Other elements increased H3.3 enrichment and displayed enhancer activity in settings of injury- and/or Neuregulin1-elicited CM proliferation. Dozens of consensus sequence motifs containing predicted transcription factor binding sites were enriched in genomic regions with regeneration-responsive H3.3 occupancy. Thus, cell-type-specific regulatory programs of tissue regeneration can be revealed by genome-wide H3.3 profiling.


Asunto(s)
Corazón/fisiología , Histonas/metabolismo , Regeneración/fisiología , Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Sitios de Unión , Elementos de Facilitación Genéticos/genética , Regulación del Desarrollo de la Expresión Génica , Histonas/genética , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Motivos de Nucleótidos/genética , Regeneración/genética , Factores de Transcripción/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
11.
Dev Cell ; 42(6): 600-615.e4, 2017 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-28950101

RESUMEN

Mechanisms that control cell-cycle dynamics during tissue regeneration require elucidation. Here we find in zebrafish that regeneration of the epicardium, the mesothelial covering of the heart, is mediated by two phenotypically distinct epicardial cell subpopulations. These include a front of large, multinucleate leader cells, trailed by follower cells that divide to produce small, mononucleate daughters. By using live imaging of cell-cycle dynamics, we show that leader cells form by spatiotemporally regulated endoreplication, caused primarily by cytokinesis failure. Leader cells display greater velocities and mechanical tension within the epicardial tissue sheet, and experimentally induced tension anisotropy stimulates ectopic endoreplication. Unbalancing epicardial cell-cycle dynamics with chemical modulators indicated autonomous regenerative capacity in both leader and follower cells, with leaders displaying an enhanced capacity for surface coverage. Our findings provide evidence that mechanical tension can regulate cell-cycle dynamics in regenerating tissue, stratifying the source cell features to improve repair.


Asunto(s)
Endorreduplicación , Pericardio/fisiología , Regeneración , Animales , Fenómenos Biomecánicos , Movimiento Celular , Células Gigantes/patología , Hipertrofia , Ratones Endogámicos C57BL , Mitosis , Poliploidía , Pez Cebra
12.
Science ; 354(6312): 630-634, 2016 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-27811277

RESUMEN

Unlike mammals, zebrafish efficiently regenerate functional nervous system tissue after major spinal cord injury. Whereas glial scarring presents a roadblock for mammalian spinal cord repair, glial cells in zebrafish form a bridge across severed spinal cord tissue and facilitate regeneration. We performed a genome-wide profiling screen for secreted factors that are up-regulated during zebrafish spinal cord regeneration. We found that connective tissue growth factor a (ctgfa) is induced in and around glial cells that participate in initial bridging events. Mutations in ctgfa disrupted spinal cord repair, and transgenic ctgfa overexpression or local delivery of human CTGF recombinant protein accelerated bridging and functional regeneration. Our study reveals that CTGF is necessary and sufficient to stimulate glial bridging and natural spinal cord regeneration.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/fisiología , Neuroglía/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Regeneración de la Medula Espinal , Proteínas de Pez Cebra/fisiología , Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente , Factor de Crecimiento del Tejido Conjuntivo/genética , Femenino , Masculino , Mutación , Regeneración de la Medula Espinal/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
13.
Elife ; 42015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25830562

RESUMEN

Heart regeneration is limited in adult mammals but occurs naturally in adult zebrafish through the activation of cardiomyocyte division. Several components of the cardiac injury microenvironment have been identified, yet no factor on its own is known to stimulate overt myocardial hyperplasia in a mature, uninjured animal. In this study, we find evidence that Neuregulin1 (Nrg1), previously shown to have mitogenic effects on mammalian cardiomyocytes, is sharply induced in perivascular cells after injury to the adult zebrafish heart. Inhibition of Erbb2, an Nrg1 co-receptor, disrupts cardiomyocyte proliferation in response to injury, whereas myocardial Nrg1 overexpression enhances this proliferation. In uninjured zebrafish, the reactivation of Nrg1 expression induces cardiomyocyte dedifferentiation, overt muscle hyperplasia, epicardial activation, increased vascularization, and causes cardiomegaly through persistent addition of wall myocardium. Our findings identify Nrg1 as a potent, induced mitogen for the endogenous adult heart regeneration program.


Asunto(s)
Corazón/fisiología , Mitógenos/metabolismo , Neurregulina-1/metabolismo , Regeneración , Proteínas de Pez Cebra/metabolismo , Pez Cebra/fisiología , Animales , Cardiomegalia/patología , Proliferación Celular , Ecocardiografía , Ventrículos Cardíacos/patología , Hiperplasia , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA