Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Cell Rep ; 43(3): 113955, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38507414

RESUMEN

Epicardial adipose tissue (eAT) is a metabolically active fat depot that has been associated with a wide array of cardiac homeostatic functions and cardiometabolic diseases. A full understanding of its diverse physiological and pathological roles is hindered by the dearth of animal models. Here, we show, in the heart of an ectothermic teleost, the zebrafish, the existence of a fat depot localized underneath the epicardium, originating from the epicardium and exhibiting the molecular signature of beige adipocytes. Moreover, a subset of adipocytes within this cardiac fat tissue exhibits primitive thermogenic potential. Transcriptomic profiling and cross-species analysis revealed elevated glycolytic and cardiac homeostatic gene expression with downregulated obesity and inflammatory hallmarks in the teleost eAT compared to that of lean aged humans. Our findings unveil epicardium-derived beige fat in the heart of an ectotherm considered to possess solely white adipocytes for energy storage and identify pathways that may underlie age-driven remodeling of human eAT.


Asunto(s)
Tejido Adiposo Beige , Pez Cebra , Animales , Humanos , Anciano , Tejido Adiposo Beige/metabolismo , Tejido Adiposo Epicárdico , Tejido Adiposo/metabolismo , Pericardio/metabolismo , Termogénesis , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo
3.
Neuron ; 112(7): 1150-1164.e6, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38295792

RESUMEN

Animals constantly need to judge the valence of an object in their environment: is it potential food or a threat? The brain makes fundamental decisions on the appropriate behavioral strategy by integrating external information from sensory organs and internal signals related to physiological needs. For example, a hungry animal may take more risks than a satiated one when deciding to approach or avoid an object. Using a proteomic profiling approach, we identified the Calmodulin-interacting peptide Pcp4a as a key regulator of foraging-related decisions. Food intake reduced abundance of protein and mRNA of pcp4a via dopamine D2-like receptor-mediated repression of adenylate cyclase. Accordingly, deleting the pcp4a gene made zebrafish larvae more risk averse in a binary decision assay. Strikingly, neurons in the tectum became less responsive to prey-like visual stimuli in pcp4a mutants, thus biasing the behavior toward avoidance. This study pinpoints a molecular mechanism modulating behavioral choice according to internal state.


Asunto(s)
Calmodulina , Pez Cebra , Animales , Pez Cebra/fisiología , Calmodulina/metabolismo , Proteómica , Neuronas/fisiología , Hambre/fisiología , Conducta Alimentaria/fisiología
4.
Dev Cell ; 58(22): 2460-2476.e7, 2023 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-37875117

RESUMEN

The autonomic nervous system plays a pivotal role in cardiac repair. Here, we describe the mechanistic underpinning of adrenergic signaling in fibrotic and regenerative response of the heart to be dependent on immunomodulation. A pharmacological approach identified adrenergic receptor alpha-1 as a key regulator of macrophage phenotypic diversification following myocardial damage in zebrafish. Genetic manipulation and single-cell transcriptomics showed that the receptor signals activation of an "extracellular matrix remodeling" transcriptional program in a macrophage subset, which serves as a key regulator of matrix composition and turnover. Mechanistically, adrenergic receptor alpha-1-activated macrophages determine activation of collagen-12-expressing fibroblasts, a cellular determinant of cardiac regenerative niche, through midkine-mediated paracrine crosstalk, allowing lymphatic and blood vessel growth and cardiomyocyte proliferation at the lesion site. These findings identify the mechanism of adrenergic signaling in macrophage phenotypic and functional determination and highlight the potential of neural modulation for regulation of fibrosis and coordination of myocardial regenerative response.


Asunto(s)
Adrenérgicos , Pez Cebra , Animales , Pez Cebra/genética , Miocardio/patología , Matriz Extracelular/patología , Macrófagos/patología , Fibrosis , Fibroblastos/patología , Receptores Adrenérgicos , Miocitos Cardíacos/patología
5.
FEBS J ; 290(4): 913-930, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-34894086

RESUMEN

Development of the heart, from early morphogenesis to functional maturation, as well as maintenance of its homeostasis are tasks requiring collaborative efforts of cardiac tissue and different extra-cardiac organ systems. The brain, lymphoid organs, and gut are among the interaction partners that can communicate with the heart through a wide array of paracrine signals acting at local or systemic level. Disturbance of cardiac homeostasis following ischemic injury also needs immediate response from these distant organs. Our hearts replace dead muscles with non-contractile fibrotic scars. We have learned from animal models capable of scarless repair that regenerative capability of the heart does not depend only on competency of the myocardium and cardiac-intrinsic factors but also on long-range molecular signals originating in other parts of the body. Here, we provide an overview of inter-organ signals that take part in development and regeneration of the heart. We highlight recent findings and remaining questions. Finally, we discuss the potential of inter-organ modulatory approaches for possible therapeutic use.


Asunto(s)
Miocitos Cardíacos , Regeneración , Animales , Regeneración/fisiología , Miocardio , Comunicación Paracrina , Modelos Animales
6.
Cardiovasc Res ; 119(5): 1234-1249, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-36536484

RESUMEN

AIMS: Dysregulated immune response contributes to inefficiency of treatment strategies to control hypertension and reduce the risk of end-organ damage. Uncovering the immune pathways driving the transition from the onset of hypertensive stimulus to the manifestation of multi-organ dysfunction are much-needed insights for immune targeted therapy. METHODS AND RESULTS: To aid visualization of cellular events orchestrating multi-organ pathogenesis, we modelled hypertensive cardiovascular remodelling in zebrafish. Zebrafish larvae exposed to ion-poor environment exhibited rapid angiotensinogen up-regulation, followed by manifestation of arterial hypertension and cardiac remodelling that recapitulates key characteristics of incipient heart failure with preserved ejection fraction. In the brain, time-lapse imaging revealed the occurrence of cerebrovascular regression through endothelial retraction and migration in response to the ion-poor treatment. This phenomenon is associated with macrophage/microglia-endothelial contacts and endothelial junctional retraction. Cytokine and transcriptomic profiling identified systemic up-regulation of interferon-γ and interleukin 1ß and revealed altered macrophage/microglia transcriptional programme characterized by suppression of innate immunity and vasculo/neuroprotective gene expression. Both zebrafish and a murine model of pressure overload-induced brain damage demonstrated that the brain pathology and macrophage/microglia phenotypic alteration are dependent on interferon-γ signalling. In zebrafish, interferon-γ receptor 1 mutation prevents cerebrovascular remodelling and dysregulation of macrophage/microglia transcriptomic profile. Supplementation of bone morphogenetic protein 5 identified from the transcriptomic approach as a down-regulated gene in ion-poor-treated macrophages/microglia that is rescued by interferon-γ blockage, mitigated cerebral microvessel loss. In mice subjected to transverse aortic constriction-induced pressure overload, typically developing cerebrovascular injury, neuroinflammation, and cognitive dysfunction, interferon-γ neutralization protected them from blood-brain barrier disruption, cerebrovascular rarefaction, and cognitive decline. CONCLUSIONS: These findings uncover cellular and molecular players of an immune pathway communicating hypertensive stimulus to structural and functional remodelling of the brain and identify anti-interferon-γ treatment as a promising intervention strategy capable of preventing pressure overload-induced damage of the cerebrovascular and nervous systems.


Asunto(s)
Disfunción Cognitiva , Hipertensión , Ratones , Animales , Pez Cebra/metabolismo , Modelos Animales de Enfermedad , Macrófagos/metabolismo , Interferón gamma/metabolismo
7.
STAR Protoc ; 3(4): 101731, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36183255

RESUMEN

The larval zebrafish has emerged as a very useful model organism to study the neuronal circuits controlling neuroendocrine and behavioral responses to stress. This protocol describes how to expose zebrafish larvae to hyperosmotic stress and test whether candidate populations of neurons are activated or inhibited by the stressor using a relatively rapid immunofluorescence staining approach. This approach takes advantage of the phosphorylation of the extracellular signal-regulated kinase (ERK) upon neuronal activation. For complete details on the use and execution of this protocol, please refer to Corradi et al. (2022).


Asunto(s)
Neuronas , Pez Cebra , Animales , Pez Cebra/fisiología , Larva , Fosforilación
8.
Curr Biol ; 32(7): 1497-1510.e5, 2022 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-35219430

RESUMEN

Animals possess neuronal circuits inducing stress to avoid or cope with threats present in their surroundings, for instance, by promoting behaviors, such as avoidance and escape. However, mechanisms must exist to tightly control responses to stressors, since overactivation of stress circuits is deleterious for the wellbeing of an organism. The underlying neuronal dynamics responsible for controlling behavioral responses to stress have remained unclear. Here, we describe a neuronal circuit in the hypothalamus of zebrafish larvae that inhibits stress-related behaviors and prevents excessive activation of the neuroendocrine pathway hypothalamic-pituitary-interrenal axis. Central components of this circuit are neurons secreting the neuropeptide Galanin, as ablation of these neurons led to abnormally high levels of stress. Surprisingly, we found that Galanin has a self-inhibitory action on Galanin-producing neurons. Our results suggest that hypothalamic Galanin-producing neurons play an important role in fine-tuning stress responses by preventing potentially harmful overactivation of stress-regulating circuits.


Asunto(s)
Galanina , Hormonas Peptídicas , Animales , Galanina/metabolismo , Hipotálamo/metabolismo , Neuronas/fisiología , Hormonas Peptídicas/metabolismo , Pez Cebra/metabolismo
9.
Cell Rep ; 33(7): 108404, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33207196

RESUMEN

Stressful experiences early in life can increase the risk of cardiovascular diseases. However, it remains largely unknown how stress influences susceptibility to the disease onset. Here, we show that exposure to brain-processed stress disrupts myocardial growth by reducing cardiomyocyte mitotic activity. Activation of the glucocorticoid receptor (GR), the primary stress response pathway, reduces cardiomyocyte numbers, disrupts trabecular formation, and leads to contractile dysfunction of the developing myocardium. However, a physiological level of GR signaling is required to prevent cardiomyocyte hyperproliferation. Mechanistically, we identify an antagonistic interaction between the GR and the cytokine interleukin-4 (IL-4) as a key player in cardiac development. IL-4 signals transcription of key regulators of cell-cycle progression in cardiomyocytes via signal transducer and activator of transcription 3 (Stat3). GR, on the contrary, inhibits this signaling system. Thus, our findings uncover an interplay between stress and immune signaling pathways critical to orchestrating physiological growth of the heart.


Asunto(s)
Interleucina-4 , Miocitos Cardíacos , Receptores de Glucocorticoides , Estrés Fisiológico , Animales , Femenino , Masculino , Ratones , Glucocorticoides/metabolismo , Interleucina-4/inmunología , Interleucina-4/metabolismo , Interleucina-4/fisiología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mitosis , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Receptores de Glucocorticoides/inmunología , Receptores de Glucocorticoides/metabolismo , Transducción de Señal/fisiología , Factor de Transcripción STAT3/metabolismo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
10.
Nat Commun ; 8(1): 1525, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29142194

RESUMEN

Cells can sacrifice their individuality by fusing, but the prevalence and significance of this process are poorly understood. To approach these questions, here we generate transgenic reporter lines in zebrafish to label and specifically ablate fused cells. In addition to skeletal muscle cells, the reporters label cardiomyocytes starting at an early developmental stage. Genetic mosaics generated by cell transplantation show cardiomyocytes expressing both donor- and host-derived transgenes, confirming the occurrence of fusion in larval hearts. These fusion events are transient and do not generate multinucleated cardiomyocytes. Functionally, cardiomyocyte fusion correlates with their mitotic activity during development as well as during regeneration in adult animals. By analyzing the cell fusion-compromised jam3b mutants, we propose a role for membrane fusion in cardiomyocyte proliferation and cardiac function. Together, our findings uncover the previously unrecognized process of transient cardiomyocyte fusion and identify its potential role in cardiac development and function.


Asunto(s)
Fusión Celular , Regulación del Desarrollo de la Expresión Génica , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas de Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Diferenciación Celular/genética , Proliferación Celular/genética , Corazón/embriología , Corazón/crecimiento & desarrollo , Larva/genética , Larva/crecimiento & desarrollo , Larva/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Miocardio/citología , Miocitos Cardíacos/citología , Pez Cebra , Proteínas de Pez Cebra/metabolismo
11.
Dev Cell ; 31(5): 640-53, 2014 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-25490269

RESUMEN

Vertebrate hematopoietic stem cells (HSCs) emerge in the aorta-gonad-mesonephros (AGM) region from "hemogenic" endothelium. Here we show that the proinflammatory cytokine interferon-γ (IFN-γ) and its receptor Crfb17 positively regulate HSC development in zebrafish. This regulation does not appear to modulate the proliferation or survival of HSCs or endothelial cells, but rather the endothelial-to-HSC transition. Notch signaling and blood flow positively regulate the expression of ifng and crfb17 in the AGM. Notably, IFN-γ overexpression partially rescues the HSC loss observed in the absence of blood flow or Notch signaling. Importantly, IFN-γ signaling acts cell autonomously to control the endothelial-to-HSC transition. IFN-γ activates Stat3, an atypical transducer of IFN-γ signaling, in the AGM, and Stat3 inhibition decreases HSC formation. Together, our findings uncover a developmental role for an inflammatory cytokine and place its action downstream of Notch signaling and blood flow to control Stat3 activation and HSC emergence.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Interferón gamma/metabolismo , Mesonefro/citología , Transducción de Señal , Pez Cebra/metabolismo , Animales , Aorta/metabolismo , Linaje de la Célula , Transducción de Señal/fisiología , Pez Cebra/embriología , Pez Cebra/genética
13.
Nat Protoc ; 5(10): 1659-65, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20885378

RESUMEN

This protocol details a culture technique for neonatal mouse retina that allows the assessment and quantification of acute responses of developing blood vessels to pharmacological manipulation. The technique has proven to be a useful tool for elucidating the molecular mechanisms that underlie the guidance of tip cells in the complex scenario of the angiogenic sprouting process. This culture setting allows the acute stimulation or inhibition of cellular functions of endothelial cells in their physiological environment ex vivo. Compared with other existing techniques, such as retinal injections in animals, the explant culture described here is an easily manageable and highly flexible alternative that allows pharmacological manipulations of the developing retina vessels. The technique involves swift extraction of retina from intact eye and retinal flat mounting on a hydrophilic membrane with minimum disturbance of the tissue. The responses of tip endothelial cell sprouting activity and filopodial extension to different angiogenic and angioinhibitory factors can be evaluated within only 4 h. The whole process for the retinal explant cultures and stimulation can be completed in 10 h.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Retina/crecimiento & desarrollo , Vasos Retinianos/crecimiento & desarrollo , Técnicas de Cultivo de Tejidos/métodos , Moduladores de la Angiogénesis , Animales , Ratones , Ratones Endogámicos C57BL , Retina/citología , Vasos Retinianos/efectos de los fármacos , Factores de Tiempo
14.
Nature ; 465(7297): 487-91, 2010 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-20445540

RESUMEN

The formation and guidance of specialized endothelial tip cells is essential for both developmental and pathological angiogenesis. Notch-1 signalling regulates the generation of tip cells, which respond to gradients of vascular endothelial growth factor (VEGF-A). The molecular cues and signalling pathways that control the guidance of tip cells are poorly understood. Bidirectional signalling by Eph receptors and ephrin ligands represents one of the most important guidance cues involved in axon path finding. Here we show that ephrin-B2 reverse signalling involving PDZ interactions regulates endothelial tip cell guidance to control angiogenic sprouting and branching in physiological and pathological angiogenesis. In vivo, ephrin-B2 PDZ-signalling-deficient mice (ephrin-B2DeltaV) exhibit a reduced number of tip cells with fewer filopodial extensions at the vascular front in the mouse retina. In pathological settings, impaired PDZ signalling decreases tumour vascularization and growth. Mechanistically, we show that ephrin-B2 controls VEGF receptor (VEGFR)-2 internalization and signalling. Importantly, internalization of VEGFR2 is necessary for activation and downstream signalling of the receptor and is required for VEGF-induced tip cell filopodial extension. Together, our results suggest that ephrin-B2 at the tip cell filopodia regulates the proper spatial activation of VEGFR2 endocytosis and signalling to direct filopodial extension. Blocking ephrin-B2 reverse signalling may be an attractive alternative or combinatorial anti-angiogenic therapy strategy to disrupt VEGFR2 function in tumour angiogenesis.


Asunto(s)
Astrocitoma/irrigación sanguínea , Astrocitoma/metabolismo , Efrina-B2/metabolismo , Neovascularización Patológica , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Astrocitoma/patología , Encéfalo/irrigación sanguínea , Células Cultivadas , Endocitosis , Células Endoteliales/citología , Células Endoteliales/metabolismo , Efrina-B2/deficiencia , Efrina-B2/genética , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Neovascularización Fisiológica , Seudópodos/metabolismo , Retina , Vasos Retinianos/citología , Vasos Retinianos/fisiología , Transducción de Señal
15.
Biochim Biophys Acta ; 1720(1-2): 44-51, 2005 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-16325760

RESUMEN

Organic anion transporter 1 (OAT1) is localized in the basolateral membrane of the proximal tubule in the kidney and plays an essential role in eliminating a wide range of organic anions, preventing their toxic effects on the body. Structural and functional studies of the transporter would be greatly assisted by inexpensive and rapid expression in the yeast Saccharomyces cerevisiae. The gene encoding rat OAT1 (rOAT1) contains many yeast non-preferred codons at the N-terminus and so was modified by fusion of the favored codon sequence of a hemagglutinin (HA) epitope preceding the start codon. The modified gene was cloned into several yeast expression plasmids, both integrative and multicopy, with either ADH1 promoter or GAL1 promoter in order to find a suitable expression system. Compared with the wild type gene, a substantial increase in rOAT1 expression was achieved by modification in the translational initiation region, suggesting that the codon chosen at the N-terminus influenced its expression. The highest inducible expression of rOAT1 was obtained under GAL1 promoter in 2 mu plasmid. A large fraction of rOAT1 was glycosylated in yeast, unaffected by growth temperature. The recombinant yeast expressing rOAT1 showed an increase in the uptake of p-aminohippurate (PAH) and this showed a positive correlation with rOAT1 expression level. Location of rOAT1 predominantly in the yeast plasma membrane confirmed correct processing. The importance of glycosylation for rOAT1 targeting was also shown. To our knowledge, this is the first successful functional expression of rOAT1 in the yeast S. cerevisiae.


Asunto(s)
Proteína 1 de Transporte de Anión Orgánico/biosíntesis , Saccharomyces cerevisiae/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Membrana Celular/metabolismo , Codón , Glicosilación , Datos de Secuencia Molecular , Proteína 1 de Transporte de Anión Orgánico/genética , Ratas , Proteínas Recombinantes/biosíntesis , Saccharomyces cerevisiae/crecimiento & desarrollo , Temperatura , Ácido p-Aminohipúrico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA