Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Exp Hematol ; 136: 104257, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38897373

RESUMEN

Hematopoietic stem cells (HSCs) possess the ability to sustain the continuous production of all blood cell types throughout an organism's lifespan. Although primarily located in the bone marrow of adults, HSCs originate during embryonic development. Visualization of the birth of HSCs, their developmental trajectory, and the specific interactions with their successive niches have significantly contributed to our understanding of the biology and mechanics governing HSC formation and expansion. Intravital techniques applied to live embryos or non-fixed samples have remarkably provided invaluable insights into the cellular and anatomical origins of HSCs. These imaging technologies have also shed light on the dynamic interactions between HSCs and neighboring cell types within the surrounding microenvironment or niche, such as endothelial cells or macrophages. This review delves into the advancements made in understanding the origin, production, and cellular interactions of HSCs, particularly during the embryonic development of mice and zebrafish, focusing on studies employing (live) imaging analysis.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas , Pez Cebra , Animales , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Pez Cebra/embriología , Ratones , Desarrollo Embrionario , Humanos , Embrión de Mamíferos/citología
2.
Cell Rep ; 39(11): 110957, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35705037

RESUMEN

Hematopoietic stem cells (HSCs) express a large variety of cell surface receptors that are associated with acquisition of self-renewal and multipotent properties. Correct expression of these receptors depends on a delicate balance between cell surface trafficking, recycling, and degradation and is controlled by the microtubule network and Golgi apparatus, whose roles have hardly been explored during embryonic/fetal hematopoiesis. Here we show that, in the absence of CLASP2, a microtubule-associated protein, the overall production of HSCs is reduced, and the produced HSCs fail to self-renew and maintain their stemness throughout mouse and zebrafish development. This phenotype can be attributed to decreased cell surface expression of the hematopoietic receptor c-Kit, which originates from increased lysosomal degradation in combination with a reduction in trafficking to the plasma membrane. A dysfunctional Golgi apparatus in CLASP2-deficient HSCs seems to be the underlying cause of the c-Kit expression and signaling imbalance.


Asunto(s)
Células Madre Hematopoyéticas , Pez Cebra , Animales , Ratones , Hematopoyesis/genética , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo
3.
Front Immunol ; 12: 790379, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34899758

RESUMEN

The journey of a hematopoietic stem cell (HSC) involves the passage through successive anatomical sites where HSCs are in direct contact with their surrounding microenvironment, also known as niche. These spatial and temporal cellular interactions throughout development are required for the acquisition of stem cell properties, and for maintaining the HSC pool through balancing self-renewal, quiescence and lineage commitment. Understanding the context and consequences of these interactions will be imperative for our understanding of HSC biology and will lead to the improvement of in vitro production of HSCs for clinical purposes. The aorta-gonad-mesonephros (AGM) region is in this light of particular interest since this is the cradle of HSC emergence during the embryonic development of all vertebrate species. In this review, we will focus on the developmental origin of HSCs and will discuss the novel technological approaches and recent progress made to identify the cellular composition of the HSC supportive niche and the underlying molecular events occurring in the AGM region.


Asunto(s)
Genómica/tendencias , Hematopoyesis/genética , Células Madre Hematopoyéticas/fisiología , Análisis de la Célula Individual/tendencias , Nicho de Células Madre , Animales , Aorta/embriología , Técnicas de Cultivo de Célula/tendencias , Linaje de la Célula , Células Cultivadas , Difusión de Innovaciones , Perfilación de la Expresión Génica/tendencias , Regulación del Desarrollo de la Expresión Génica , Gónadas/embriología , Humanos , Mesonefro/embriología , Fenotipo , Proteómica/tendencias , Transducción de Señal , Transcriptoma
4.
Nat Cell Biol ; 23(4): 322-329, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33837285

RESUMEN

De novo blood vessel formation occurs through coalescence of endothelial cells (ECs) into a cord-like structure, followed by lumenization either through cell-1-3 or cord-hollowing4-7. Vessels generated in this manner are restricted in diameter to one or two ECs, and these models fail to explain how vasculogenesis can form large-diameter vessels. Here, we describe a model for large vessel formation that does not require a cord-like structure or a hollowing step. In this model, ECs coalesce into a network of struts in the future lumen of the vessel, a process dependent upon bone morphogenetic protein signalling. The vessel wall forms around this network and consists initially of only a few patches of ECs. To withstand external forces and to maintain the shape of the vessel, strut formation traps erythrocytes into compartments to form a rigid structure. Struts gradually prune and ECs from struts migrate into and become part of the vessel wall. Experimental severing of struts resulted in vessel collapse, disturbed blood flow and remodelling defects, demonstrating that struts enable the patency of large vessels during their formation.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Células Endoteliales/fisiología , Morfogénesis/genética , Neovascularización Fisiológica/genética , Vasos Sanguíneos/metabolismo , Células Endoteliales/metabolismo , Eritrocitos/metabolismo , Eritrocitos/patología , Humanos
5.
Blood ; 136(7): 831-844, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32457985

RESUMEN

The defined location of a stem cell within a niche regulates its fate, behavior, and molecular identity via a complex extrinsic regulation that is far from being fully elucidated. To explore the molecular characteristics and key components of the aortic microenvironment, where the first hematopoietic stem cells are generated during development, we performed genome-wide RNA tomography sequencing on zebrafish, chicken, mouse, and human embryos. The resulting anterior-posterior and dorsal-ventral transcriptional maps provided a powerful resource for exploring genes and regulatory pathways active in the aortic microenvironment. By performing interspecies comparative RNA sequencing analyses and functional assays, we explored the complexity of the aortic microenvironment landscape and the fine-tuning of various factors interacting to control hematopoietic stem cell generation, both in time and space in vivo, including the ligand-receptor couple ADM-RAMP2 and SVEP1. Understanding the regulatory function of the local environment will pave the way for improved stem cell production in vitro and clinical cell therapy.


Asunto(s)
Aorta/embriología , Células Madre Hematopoyéticas/citología , ARN/análisis , Nicho de Células Madre/genética , Tomografía , Animales , Animales Modificados Genéticamente , Aorta/citología , Rastreo Celular/métodos , Embrión de Pollo , Embrión de Mamíferos , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , ARN/genética , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual , Especificidad de la Especie , Tomografía/métodos , Tomografía/veterinaria , Pez Cebra/embriología , Pez Cebra/genética
6.
Nat Commun ; 9(1): 5314, 2018 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-30552331

RESUMEN

Arteries and veins are formed independently by different types of endothelial cells (ECs). In vascular remodeling, arteries and veins become connected and some arteries become veins. It is unclear how ECs in transforming vessels change their type and how fates of individual vessels are determined. In embryonic zebrafish trunk, vascular remodeling transforms arterial intersegmental vessels (ISVs) into a functional network of arteries and veins. Here we find that, once an ISV is connected to venous circulation, venous blood flow promotes upstream migration of ECs that results in displacement of arterial ECs by venous ECs, completing the transformation of this ISV into a vein without trans-differentiation of ECs. Arterial blood flow initiated in two neighboring ISVs prevents their transformation into veins by activating Notch signaling in ECs. Together, different responses of ECs to arterial and venous blood flow lead to formation of a balanced network with equal numbers of arteries and veins.


Asunto(s)
Arterias/citología , Arterias/embriología , Células Endoteliales/citología , Células Endoteliales/fisiología , Remodelación Vascular/fisiología , Venas/citología , Venas/embriología , Pez Cebra/embriología , Animales , Diferenciación Celular/fisiología , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Morfolinos , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
7.
Trends Cell Biol ; 28(1): 58-66, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28882414

RESUMEN

Hematopoietic stem cells (HSCs) have the extraordinary ability to both self-renew and generate all mature blood cell lineages. The ability to produce or expand patient-derived HSCs in vitro would greatly improve the outcome for patients with blood disorders that are currently treated with allogeneic HSC transplantation. Many laboratories have been working to identify the signals required for HSC emergence in their native environments to apply this knowledge in vitro. Recently, several signals traditionally known to underlie classical inflammation have emerged as essential regulators of HSC development. In this review we synthesize the findings that have established inflammatory cues as key regulators of HSC development.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Inflamación/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula/fisiología , Citocinas/metabolismo , Hematopoyesis Extramedular/fisiología , Humanos , Transducción de Señal
8.
Zebrafish ; 14(5): 489-491, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28118101

RESUMEN

We describe the design, fabrication, and applications of a four-well dish for imaging of the trunk of larval zebrafish. The dish facilitates immobilization of anesthetized zebrafish larvae, with their tails gently pushed against a microscope cover glass, enabling longitudinal imaging at 24-72 h postfertilization using high-resolution objective lenses.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Inmovilización/instrumentación , Microscopía Confocal/métodos , Pez Cebra/anatomía & histología , Pez Cebra/crecimiento & desarrollo , Animales , Técnicas de Cultivo de Célula , Larva/anatomía & histología , Larva/ultraestructura
9.
Development ; 142(6): 1050-61, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25758220

RESUMEN

The adult blood system is established by hematopoietic stem cells (HSCs), which arise during development from an endothelial-to-hematopoietic transition of cells comprising the floor of the dorsal aorta. Expression of aortic runx1 has served as an early marker of HSC commitment in the zebrafish embryo, but recent studies have suggested that HSC specification begins during the convergence of posterior lateral plate mesoderm (PLM), well before aorta formation and runx1 transcription. Further understanding of the earliest stages of HSC specification necessitates an earlier marker of hemogenic endothelium. Studies in mice have suggested that GATA2 might function at early stages within hemogenic endothelium. Two orthologs of Gata2 exist in zebrafish: gata2a and gata2b. Here, we report that gata2b expression initiates during the convergence of PLM, becoming restricted to emerging HSCs. We observe Notch-dependent gata2b expression within the hemogenic subcompartment of the dorsal aorta that is in turn required to initiate runx1 expression. Our results indicate that Gata2b functions within hemogenic endothelium from an early stage, whereas Gata2a functions more broadly throughout the vascular system.


Asunto(s)
Tipificación del Cuerpo/fisiología , Factor de Transcripción GATA2/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Hemangioblastos/fisiología , Proteínas de Pez Cebra/genética , Pez Cebra/embriología , Animales , Aorta/citología , Aorta/embriología , Proteínas Bacterianas , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Cartilla de ADN/genética , Citometría de Flujo , Factor de Transcripción GATA2/genética , Regulación del Desarrollo de la Expresión Génica/genética , Hibridación in Situ , Proteínas Luminiscentes , Mesodermo/embriología , Oligonucleótidos Antisentido/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Imagen de Lapso de Tiempo , Proteínas de Pez Cebra/metabolismo , Proteína Fluorescente Roja
10.
Dev Cell ; 27(5): 574-85, 2013 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-24290981

RESUMEN

Epithelial cell migration is crucial for the development and regeneration of epithelial tissues. Aberrant regulation of epithelial cell migration has a major role in pathological processes such as the development of cancer metastasis and tissue fibrosis. Here, we report that in response to factors that promote cell motility, the Rap guanine exchange factor RAPGEF2 is rapidly phosphorylated by I-kappa-B-kinase-ß and casein kinase-1α and consequently degraded by the proteasome via the SCF(ßTrCP) ubiquitin ligase. Failure to degrade RAPGEF2 in epithelial cells results in sustained activity of Rap1 and inhibition of cell migration induced by HGF, a potent metastatic factor. Furthermore, expression of a degradation-resistant RAPGEF2 mutant greatly suppresses dissemination and metastasis of human breast cancer cells. These findings reveal a molecular mechanism regulating migration and invasion of epithelial cells and establish a key direct link between IKKß and cell motility controlled by Rap-integrin signaling.


Asunto(s)
Caseína Quinasa Ialfa/metabolismo , Movimiento Celular/fisiología , Células Epiteliales/citología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Quinasa I-kappa B/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal/fisiología , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Neoplasias de la Mama , Línea Celular Tumoral , Femenino , Células HEK293 , Xenoinjertos , Humanos , Masculino , Fosforilación/fisiología , Proteínas Ligasas SKP Cullina F-box/metabolismo , Pez Cebra
11.
PLoS One ; 8(9): e73693, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24069224

RESUMEN

Lymphatic vessels are derived from venous endothelial cells and their formation is governed by the Vascular endothelial growth factor C (VegfC)/Vegf receptor 3 (Vegfr3; Flt4) signaling pathway. Recent studies show that Collagen and Calcium Binding EGF domains 1 protein (Ccbe1) enhances VegfC-dependent lymphangiogenesis. Both Ccbe1 and Flt4 have been shown to be indispensable for lymphangiogenesis. However, how these essential players are transcriptionally regulated remains poorly understood. In the case of angiogenesis, atypical E2fs (E2f7 and E2f8) however have been recently shown to function as transcriptional activators for VegfA. Using a genome-wide approach we here identified both CCBE1 and FLT4 as direct targets of atypical E2Fs. E2F7/8 directly bind and stimulate the CCBE1 promoter, while recruitment of E2F7/8 inhibits the FLT4 promoter. Importantly, inactivation of e2f7/8 in zebrafish impaired venous sprouting and lymphangiogenesis with reduced ccbe1 expression and increased flt4 expression. Remarkably, over-expression of e2f7/8 rescued Ccbe1- and Flt4-dependent lymphangiogenesis phenotypes. Together these results identified E2f7/8 as novel in vivo transcriptional regulators of Ccbe1 and Flt4, both essential genes for venous sprouting and lymphangiogenesis.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Linfangiogénesis/fisiología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Western Blotting , Proteínas de Unión al Calcio/genética , Inmunoprecipitación de Cromatina , Electroforesis en Gel de Poliacrilamida , Humanos , Linfangiogénesis/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Pez Cebra , Proteínas de Pez Cebra/genética
12.
J Pathol ; 231(1): 117-29, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23744542

RESUMEN

Biallelic mutations of the von Hippel-Lindau (VHL) gene are the most common cause of sporadic and inherited renal cell carcinoma (RCC). Loss of VHL has been reported to affect cell proliferation by deregulating cell cycle-associated proteins. We report that the VHL gene product (pVHL) inhibits E2F1 expression at both mRNA and protein level in zebrafish and human RCC cells, while loss of VHL increases E2F1 expression in patient kidney tumour tissue and RCC cells, resulting in a delay of cell cycle progression. RCCs from von Hippel-Lindau patients with known germline VHL mutations express significantly more E2F1 compared to sporadic RCCs with either clear-cell (cc) or non-cc histology. Analysis of 138 primary RCCs reveals that E2F1 expression is significantly higher in tumours with a diameter ≤7 cm and with a favourable American Joint Committee on Cancer (AJCC) stage. The expression of E2F1 in RCC significantly correlates with p27 expression, suggesting that increased expression of E2F1 in RCC induces tumour cell senescence via p27. Cox regression analysis shows significant prediction of E2F1 expression for disease-free survival and overall survival, implying that E2F1 expression in kidney tumour is a novel prognostic factor for patients with RCC.


Asunto(s)
Carcinoma de Células Renales/mortalidad , Factor de Transcripción E2F1/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Neoplasias Renales/mortalidad , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/fisiología , Animales , Western Blotting , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Senescencia Celular , Modelos Animales de Enfermedad , Factor de Transcripción E2F1/metabolismo , Femenino , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Masculino , Persona de Mediana Edad , Organismos Modificados Genéticamente , Plásmidos , Pronóstico , Antígeno Nuclear de Célula en Proliferación/metabolismo , Modelos de Riesgos Proporcionales , Reacción en Cadena en Tiempo Real de la Polimerasa , Tasa de Supervivencia , Transfección , Células Tumorales Cultivadas , Pez Cebra
13.
Dis Model Mech ; 6(5): 1159-66, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23720233

RESUMEN

Angiogenesis, the emergence of vessels from an existing vascular network, is pathologically associated with tumor progression and is of great interest for therapeutic intervention. PTEN is a frequently mutated tumor suppressor and has been linked to the progression of many types of tumors, including hemangiosarcomas in zebrafish. Here, we report that mutant zebrafish embryos lacking functional Pten exhibit enhanced angiogenesis, accompanied by elevated levels of phosphorylated Akt (pAkt). Inhibition of phosphoinositide 3-kinase (PI3K) by LY294002 treatment and application of sunitinib, a widely used anti-angiogenic compound, suppressed enhanced angiogenesis in Pten mutants. Vegfaa has a crucial role in angiogenesis and vegfaa expression was upregulated in embryos lacking functional Pten. Interestingly, vegfaa expression was also upregulated in hemangiosarcomas from haploinsufficient adult zebrafish Pten mutants. Elevated vegfaa expression in mutant embryos lacking functional Pten was suppressed by LY294002. Surprisingly, sunitinib treatment dramatically enhanced vegfaa expression in Pten mutant embryos, which might account for tumor relapse in human patients who are treated with sunitinib. Combined treatment with suboptimal concentrations of sunitinib and LY294002 rescued enhanced angiogenesis in pten mutant embryos without the dramatic increase in vegfaa expression, suggesting a new approach for therapeutic intervention in VEGFR-signaling-dependent tumors.


Asunto(s)
Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Fosfoproteínas Fosfatasas/deficiencia , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Cromonas/farmacología , Cromonas/uso terapéutico , Quimioterapia Combinada , Células Endoteliales/metabolismo , Células Endoteliales/patología , Haploinsuficiencia/efectos de los fármacos , Haploinsuficiencia/genética , Hemangiosarcoma/irrigación sanguínea , Hemangiosarcoma/tratamiento farmacológico , Hemangiosarcoma/patología , Humanos , Indoles/farmacología , Indoles/uso terapéutico , Morfolinas/farmacología , Morfolinas/uso terapéutico , Mutación/genética , Neovascularización Patológica/tratamiento farmacológico , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Pirroles/farmacología , Pirroles/uso terapéutico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Sunitinib , Regulación hacia Arriba/efectos de los fármacos , Pez Cebra/embriología , Proteínas de Pez Cebra/genética
14.
Transcription ; 4(2): 62-6, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23412359

RESUMEN

Recently, we showed that E2F7 and E2F8 (E2F7/8) are critical regulators of angiogenesis through transcriptional control of VEGFA in cooperation with HIF. (1) Here we investigate the existence of other novel putative angiogenic E2F7/8-HIF targets, and discuss the role of the RB-E2F pathway in regulating angiogenesis during embryonic and tumor development.


Asunto(s)
Factores de Transcripción E2F/metabolismo , Factor 1 Inducible por Hipoxia/metabolismo , Proteínas Represoras/metabolismo , Animales , Sitios de Unión , Factores de Transcripción E2F/deficiencia , Factores de Transcripción E2F/genética , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
EMBO J ; 31(19): 3871-84, 2012 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-22903062

RESUMEN

The E2F family of transcription factors plays an important role in controlling cell-cycle progression. While this is their best-known function, we report here novel functions for the newest members of the E2F family, E2F7 and E2F8 (E2F7/8). We show that simultaneous deletion of E2F7/8 in zebrafish and mice leads to severe vascular defects during embryonic development. Using a panel of transgenic zebrafish with fluorescent-labelled blood vessels, we demonstrate that E2F7/8 are essential for proper formation of blood vessels. Despite their classification as transcriptional repressors, we provide evidence for a molecular mechanism through which E2F7/8 activate the transcription of the vascular endothelial growth factor A (VEGFA), a key factor in guiding angiogenesis. We show that E2F7/8 directly bind and stimulate the VEGFA promoter independent of canonical E2F binding elements. Instead, E2F7/8 form a transcriptional complex with the hypoxia inducible factor 1 (HIF1) to stimulate VEGFA promoter activity. These results uncover an unexpected link between E2F7/8 and the HIF1-VEGFA pathway providing a molecular mechanism by which E2F7/8 control angiogenesis.


Asunto(s)
Factores de Transcripción E2F/metabolismo , Factor 1 Inducible por Hipoxia/metabolismo , Neovascularización Fisiológica/genética , Activación Transcripcional , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Animales Modificados Genéticamente , Línea Celular Tumoral , Factores de Transcripción E2F/genética , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Eliminación de Gen , Humanos , Ratones , Regiones Promotoras Genéticas , Pez Cebra
16.
J Biol Chem ; 283(32): 22295-303, 2008 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-18477567

RESUMEN

One major intracellular signaling pathway involved in heart failure employs the phosphatase calcineurin and its downstream transcriptional effector nuclear factor of activated T-cells (NFAT). In vivo evidence for the involvement of NFAT factors in heart failure development is still ill defined. Here we reveal that nfatc2 transcripts outnumber those from other nfat genes in the unstimulated heart by severalfold. Transgenic mice with activated calcineurin in the postnatal myocardium crossbred with nfatc2-null mice revealed a significant abrogation of calcineurin-provoked cardiac growth, indicating that NFATc2 plays an important role downstream of calcineurin and validates the original hypothesis that calcineurin mediates myocyte hypertrophy through activation of NFAT transcription factors. In the absence of NFATc2, a clear protection against the geometrical, functional, and molecular deterioration of the myocardium following biomechanical stress was also evident. In contrast, physiological cardiac enlargement in response to voluntary exercise training was not affected in nfatc2-null mice. Combined, these results reveal a major role for the NFATc2 transcription factor in pathological cardiac remodeling and heart failure.


Asunto(s)
Calcineurina/metabolismo , Cardiomegalia/fisiopatología , Insuficiencia Cardíaca/fisiopatología , Factores de Transcripción NFATC/metabolismo , Animales , Ratones , Ratones Transgénicos , Miocardio/química , Factores de Transcripción NFATC/genética , Isoformas de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA