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











Base de dados
Intervalo de ano de publicação
1.
J Biomed Opt ; 28(6): 066502, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37351197

RESUMO

Significance: Fluorescence lifetime imaging microscopy (FLIM) of the metabolic co-enzyme nicotinamide adenine dinucleotide (phosphate) [NAD(P)H] is a popular method to monitor single-cell metabolism within unperturbed, living 3D systems. However, FLIM of NAD(P)H has not been performed in a light-sheet geometry, which is advantageous for rapid imaging of cells within live 3D samples. Aim: We aim to design, validate, and demonstrate a proof-of-concept light-sheet system for NAD(P)H FLIM. Approach: A single-photon avalanche diode camera was integrated into a light-sheet microscope to achieve optical sectioning and limit out-of-focus contributions for NAD(P)H FLIM of single cells. Results: An NAD(P)H light-sheet FLIM system was built and validated with fluorescence lifetime standards and with time-course imaging of metabolic perturbations in pancreas cancer cells with 10 s integration times. NAD(P)H light-sheet FLIM in vivo was demonstrated with live neutrophil imaging in a larval zebrafish tail wound also with 10 s integration times. Finally, the theoretical and practical imaging speeds for NAD(P)H FLIM were compared across laser scanning and light-sheet geometries, indicating a 30× to 6× acquisition speed advantage for the light sheet compared to the laser scanning geometry. Conclusions: FLIM of NAD(P)H is feasible in a light-sheet geometry and is attractive for 3D live cell imaging applications, such as monitoring immune cell metabolism and migration within an organism.


Assuntos
NAD , Neoplasias Pancreáticas , Animais , NAD/metabolismo , Peixe-Zebra , Microscopia de Fluorescência/métodos , Fótons , Imagem Óptica/métodos
2.
bioRxiv ; 2023 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-36778488

RESUMO

Single photon avalanche diode (SPAD) array sensors can increase the imaging speed for fluorescence lifetime imaging microscopy (FLIM) by transitioning from laser scanning to widefield geometries. While a SPAD camera in epi-fluorescence geometry enables widefield FLIM of fluorescently labeled samples, label-free imaging of single-cell autofluorescence is not feasible in an epi-fluorescence geometry because background fluorescence from out-of-focus features masks weak cell autofluorescence and biases lifetime measurements. Here, we address this problem by integrating the SPAD camera in a light sheet illumination geometry to achieve optical sectioning and limit out-of-focus contributions, enabling fast label-free FLIM of single-cell NAD(P)H autofluorescence. The feasibility of this NAD(P)H light sheet FLIM system was confirmed with time-course imaging of metabolic perturbations in pancreas cancer cells with 10 s integration times, and in vivo NAD(P)H light sheet FLIM was demonstrated with live neutrophil imaging in a zebrafish tail wound, also with 10 s integration times. Finally, the theoretical and practical imaging speeds for NAD(P)H FLIM were compared across laser scanning and light sheet geometries, indicating a 30X to 6X frame rate advantage for the light sheet compared to the laser scanning geometry. This light sheet system provides faster frame rates for 3D NAD(P)H FLIM for live cell imaging applications such as monitoring single cell metabolism and immune cell migration throughout an entire living organism.

3.
Proc Natl Acad Sci U S A ; 119(50): e2201097119, 2022 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-36469766

RESUMO

Despite the robust healing capacity of the liver, regenerative failure underlies numerous hepatic diseases, including the JAG1 haploinsufficient disorder, Alagille syndrome (ALGS). Cholestasis due to intrahepatic duct (IHD) paucity resolves in certain ALGS cases but fails in most with no clear mechanisms or therapeutic interventions. We find that modulating jag1b and jag2b allele dosage is sufficient to stratify these distinct outcomes, which can be either exacerbated or rescued with genetic manipulation of Notch signaling, demonstrating that perturbations of Jag/Notch signaling may be causal for the spectrum of ALGS liver severities. Although regenerating IHD cells proliferate, they remain clustered in mutants that fail to recover due to a blunted elevation of Notch signaling in the distal-most IHD cells. Increased Notch signaling is required for regenerating IHD cells to branch and segregate into the peripheral region of the growing liver, where biliary paucity is commonly observed in ALGS. Mosaic loss- and-gain-of-function analysis reveals Sox9b to be a key Notch transcriptional effector required cell autonomously to regulate these cellular dynamics during IHD regeneration. Treatment with a small-molecule putative Notch agonist stimulates Sox9 expression in ALGS patient fibroblasts and enhances hepatic sox9b expression, rescues IHD paucity and cholestasis, and increases survival in zebrafish mutants, thereby providing a proof-of-concept therapeutic avenue for this disorder.


Assuntos
Síndrome de Alagille , Ductos Biliares Intra-Hepáticos , Transdução de Sinais , Animais , Humanos , Síndrome de Alagille/genética , Síndrome de Alagille/metabolismo , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Mosaicismo , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Regeneração , Ductos Biliares Intra-Hepáticos/citologia , Ductos Biliares Intra-Hepáticos/patologia , Fibroblastos
4.
J Cell Biol ; 221(8)2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35708547

RESUMO

Many cells can generate complementary traveling waves of actin filaments (F-actin) and cytoskeletal regulators. This phenomenon, termed cortical excitability, results from coupled positive and negative feedback loops of cytoskeletal regulators. The nature of these feedback loops, however, remains poorly understood. We assessed the role of the Rho GAP RGA-3/4 in the cortical excitability that accompanies cytokinesis in both frog and starfish. RGA-3/4 localizes to the cytokinetic apparatus, "chases" Rho waves in an F-actin-dependent manner, and when coexpressed with the Rho GEF Ect2, is sufficient to convert the normally quiescent, immature Xenopus oocyte cortex into a dramatically excited state. Experiments and modeling show that changing the ratio of RGA-3/4 to Ect2 produces cortical behaviors ranging from pulses to complex waves of Rho activity. We conclude that RGA-3/4, Ect2, Rho, and F-actin form the core of a versatile circuit that drives a diverse range of cortical behaviors, and we demonstrate that the immature oocyte is a powerful model for characterizing these dynamics.


Assuntos
Actinas , Citoesqueleto , Proteínas Ativadoras de GTPase , Proteínas Proto-Oncogênicas , Proteínas rho de Ligação ao GTP , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Citocinese , Citoesqueleto/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Oócitos , Proteínas Proto-Oncogênicas/metabolismo , Xenopus , Proteínas rho de Ligação ao GTP/metabolismo
5.
Nat Commun ; 13(1): 1677, 2022 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-35354817

RESUMO

The mesothelium lines body cavities and surrounds internal organs, widely contributing to homeostasis and regeneration. Mesothelium disruptions cause visceral anomalies and mesothelioma tumors. Nonetheless, the embryonic emergence of mesothelia remains incompletely understood. Here, we track mesothelial origins in the lateral plate mesoderm (LPM) using zebrafish. Single-cell transcriptomics uncovers a post-gastrulation gene expression signature centered on hand2 in distinct LPM progenitor cells. We map mesothelial progenitors to lateral-most, hand2-expressing LPM and confirm conservation in mouse. Time-lapse imaging of zebrafish hand2 reporter embryos captures mesothelium formation including pericardium, visceral, and parietal peritoneum. We find primordial germ cells migrate with the forming mesothelium as ventral migration boundary. Functionally, hand2 loss disrupts mesothelium formation with reduced progenitor cells and perturbed migration. In mouse and human mesothelioma, we document expression of LPM-associated transcription factors including Hand2, suggesting re-initiation of a developmental program. Our data connects mesothelium development to Hand2, expanding our understanding of mesothelial pathologies.


Assuntos
Mesotelioma , Peixe-Zebra , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Epitélio/metabolismo , Mesotelioma/genética , Camundongos , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
6.
Hepatology ; 75(3): 567-583, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34569629

RESUMO

BACKGROUND AND AIMS: Alagille Syndrome (ALGS) is a congenital disorder caused by mutations in the Notch ligand gene JAGGED1, leading to neonatal loss of intrahepatic duct (IHD) cells and cholestasis. Cholestasis can resolve in certain patients with ALGS, suggesting regeneration of IHD cells. However, the mechanisms driving IHD cell regeneration following Jagged loss remains unclear. Here, we show that cholestasis due to developmental loss of IHD cells can be consistently phenocopied in zebrafish with compound jagged1b and jagged2b mutations or knockdown. APPROACH AND RESULTS: Leveraging the transience of jagged knockdown in juvenile zebrafish, we find that resumption of Jagged expression leads to robust regeneration of IHD cells through a Notch-dependent mechanism. Combining multiple lineage tracing strategies with whole-liver three-dimensional imaging, we demonstrate that the extrahepatic duct (EHD) is the primary source of multipotent progenitors that contribute to the regeneration, but not to the development, of IHD cells. Hepatocyte-to-IHD cell transdifferentiation is possible but rarely detected. Progenitors in the EHD proliferate and migrate into the liver with Notch signaling loss and differentiate into IHD cells if Notch signaling increases. Tissue-specific mosaic analysis with an inducible dominant-negative Fgf receptor suggests that Fgf signaling from the surrounding mesenchymal cells maintains this extrahepatic niche by directly preventing premature differentiation and allocation of EHD progenitors to the liver. Indeed, transcriptional profiling and functional analysis of adult mouse EHD organoids uncover their distinct differentiation and proliferative potential relative to IHD organoids. CONCLUSIONS: Our data show that IHD cells regenerate upon resumption of Jagged/Notch signaling, from multipotent progenitors originating from an Fgf-dependent extrahepatic stem cell niche. We posit that if Jagged/Notch signaling is augmented, through normal stochastic variation, gene therapy, or a Notch agonist, regeneration of IHD cells in patients with ALGS may be enhanced.


Assuntos
Síndrome de Alagille , Ductos Biliares Extra-Hepáticos , Ductos Biliares Intra-Hepáticos , Proteínas de Ligação ao Cálcio , Proteína Jagged-1 , Regeneração Hepática/fisiologia , Receptores Notch/metabolismo , Proteínas de Peixe-Zebra , Síndrome de Alagille/genética , Síndrome de Alagille/metabolismo , Animais , Ductos Biliares Extra-Hepáticos/crescimento & desenvolvimento , Ductos Biliares Extra-Hepáticos/fisiologia , Ductos Biliares Intra-Hepáticos/crescimento & desenvolvimento , Ductos Biliares Intra-Hepáticos/fisiologia , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Transdiferenciação Celular , Modelos Animais de Doenças , Humanos , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Fígado/crescimento & desenvolvimento , Fígado/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
7.
Nat Commun ; 12(1): 7180, 2021 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-34893591

RESUMO

Metastasis is the main cause of carcinoma-related death, yet we know little about how it initiates due to our inability to visualize stochastic invasion events. Classical models suggest that cells accumulate mutations that first drive formation of a primary mass, and then downregulate epithelia-specific genes to cause invasion and metastasis. Here, using transparent zebrafish epidermis to model simple epithelia, we can directly image invasion. We find that KRas-transformation, implicated in early carcinogenesis steps, directly drives cell invasion by hijacking a process epithelia normally use to promote death-cell extrusion. Cells invading by basal cell extrusion simultaneously pinch off their apical epithelial determinants, endowing new plasticity. Following invasion, cells divide, enter the bloodstream, and differentiate into stromal, neuronal-like, and other cell types. Yet, only invading KRasV12 cells deficient in p53 survive and form internal masses. Together, we demonstrate that KRas-transformation alone causes cell invasion and partial dedifferentiation, independently of mass formation.


Assuntos
Células Epiteliais/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Animais , Movimento Celular , Epiderme/metabolismo , Epitélio/metabolismo , Humanos , Neoplasias/diagnóstico por imagem , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra
8.
Sci Rep ; 10(1): 13254, 2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32764590

RESUMO

Understanding mechanisms mediating tumor metastasis is crucial for diagnostic and therapeutic targeting. Here, we take advantage of a transparent embryonic zebrafish xenograft model (eZXM) to visualize and track metastatic cells in real time using selective plane illumination microscopy (SPIM) for up to 30 h. Injected human leukemic and breast cancer cells exhibited cell-type specific patterns of intravascular distribution with leukemic cells moving faster than breast cancer cells. Tracking of tumor cells from high-resolution images revealed acute differences in intravascular speed and distance covered by cells. While the majority of injected breast cancer cells predominantly adhered to nearby vasculature, about 30% invaded the non-vascularized tissue, reminiscent of their metastatic phenotype. Survival of the injected tumor cells appeared to be partially inhibited and time-lapse imaging showed a possible role for host macrophages of the recipient embryos. Leukemic cell dissemination could be effectively blocked by pharmacological ROCK1 inhibition using Fasudil. These observations, and the ability to image several embryos simultaneously, support the use of eZXM and SPIM imaging as a functional screening platform to identify compounds that suppress cancer cell spread and invasion.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , Neoplasias da Mama/diagnóstico por imagem , Leucemia/diagnóstico por imagem , Metástase Neoplásica/diagnóstico por imagem , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/administração & dosagem , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/uso terapêutico , Animais , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Rastreamento de Células , Feminino , Leucemia/tratamento farmacológico , Microscopia , Invasividade Neoplásica , Metástase Neoplásica/tratamento farmacológico , Transplante de Neoplasias , Imagem com Lapso de Tempo , Peixe-Zebra
9.
Biomed Opt Express ; 11(5): 2591-2606, 2020 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-32499946

RESUMO

High-throughput drug screening of patient-derived organoids offers an attractive platform to determine cancer treatment efficacy. Here, selective plane illumination microscopy (SPIM) was used to determine treatment response in organoids with endogenous fluorescence from the metabolic coenzymes NAD(P)H and FAD. Rapid 3-D autofluorescence imaging of colorectal cancer organoids was achieved. A quantitative image analysis approach was developed to segment each organoid and quantify changes in endogenous fluorescence caused by treatment. Quantitative analysis of SPIM volumes confirmed the sensitivity of patient-derived organoids to standard therapies. This proof-of-principle study demonstrates that SPIM is a powerful tool for high-throughput screening of organoid treatment response.

10.
J Cereb Blood Flow Metab ; 40(2): 298-313, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-30398083

RESUMO

Neurovascular coupling (through which local cerebral blood flow changes in response to neural activation are mediated) is impaired in many diseases including diabetes. Current preclinical rodent models of neurovascular coupling rely on invasive surgery and instrumentation, but transgenic zebrafish coupled with advances in imaging techniques allow non-invasive quantification of cerebrovascular anatomy, neural activation, and cerebral vessel haemodynamics. We therefore established a novel non-invasive, non-anaesthetised zebrafish larval model of neurovascular coupling, in which visual stimulus evokes neuronal activation in the optic tectum that is associated with a specific increase in red blood cell speed in tectal blood vessels. We applied this model to the examination of the effect of glucose exposure on cerebrovascular patterning and neurovascular coupling. We found that chronic exposure of zebrafish to glucose impaired tectal blood vessel patterning and neurovascular coupling. The nitric oxide donor sodium nitroprusside rescued all these adverse effects of glucose exposure on cerebrovascular patterning and function. Our results establish the first non-mammalian model of neurovascular coupling, offering the potential to perform more rapid genetic modifications and high-throughput screening than is currently possible using rodents. Furthermore, using this zebrafish model, we reveal a potential strategy to ameliorate the effects of hyperglycemia on cerebrovascular function.


Assuntos
Encéfalo , Circulação Cerebrovascular , Hiperglicemia , Neovascularização Patológica , Acoplamento Neurovascular , Potenciais de Ação , Animais , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Encéfalo/fisiopatologia , Artérias Cerebrais/patologia , Artérias Cerebrais/fisiopatologia , Veias Cerebrais/patologia , Veias Cerebrais/fisiopatologia , Hiperglicemia/sangue , Hiperglicemia/patologia , Hiperglicemia/fisiopatologia , Neovascularização Patológica/patologia , Neovascularização Patológica/fisiopatologia , Peixe-Zebra
11.
Artigo em Inglês | MEDLINE | ID: mdl-31740552

RESUMO

Antifungal therapy can fail in a remarkable number of patients with invasive fungal disease, resulting in significant morbidity worldwide. A major contributor to this failure is that while these drugs have high potency in vitro, we do not fully understand how they work inside infected hosts. Here, we used a transparent larval zebrafish model of Aspergillus fumigatus infection amenable to real-time imaging of invasive disease as an in vivo intermediate vertebrate model to investigate the efficacy and mechanism of the antifungal drug voriconazole. We found that the ability of voriconazole to protect against A. fumigatus infection depends on host innate immune cells and, specifically, on the presence of macrophages. While voriconazole inhibits fungal spore germination and growth in vitro, it does not do so in larval zebrafish. Instead, live imaging of whole, intact larvae over a multiday course of infection revealed that macrophages slow down initial fungal growth, allowing voriconazole time to target and kill A. fumigatus hyphae postgermination. These findings shed light on how antifungal drugs such as voriconazole may synergize with the immune response in living hosts.


Assuntos
Antifúngicos/uso terapêutico , Aspergilose/tratamento farmacológico , Aspergilose/imunologia , Aspergillus fumigatus , Voriconazol/uso terapêutico , Animais , Aspergilose/microbiologia , Imunidade Celular , Larva , Macrófagos/imunologia , Testes de Sensibilidade Microbiana , Esporos Fúngicos/imunologia , Peixe-Zebra
12.
EMBO Rep ; 20(8): e47047, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31379129

RESUMO

We identify a novel endothelial membrane behaviour in transgenic zebrafish. Cerebral blood vessels extrude large transient spherical structures that persist for an average of 23 min before regressing into the parent vessel. We term these structures "kugeln", after the German for sphere. Kugeln are only observed arising from the cerebral vessels and are present as late as 28 days post fertilization. Kugeln do not communicate with the vessel lumen and can form in the absence of blood flow. They contain little or no cytoplasm, but the majority are highly positive for nitric oxide reactivity. Kugeln do not interact with brain lymphatic endothelial cells (BLECs) and can form in their absence, nor do they perform a scavenging role or interact with macrophages. Inhibition of actin polymerization, Myosin II, or Notch signalling reduces kugel formation, while inhibition of VEGF or Wnt dysregulation (either inhibition or activation) increases kugel formation. Kugeln represent a novel Notch-dependent NO-containing endothelial organelle restricted to the cerebral vessels, of currently unknown function.


Assuntos
Vasos Sanguíneos/citologia , Encéfalo/citologia , Células Endoteliais/ultraestrutura , Regulação da Expressão Gênica no Desenvolvimento , Neovascularização Fisiológica/genética , Peixe-Zebra/embriologia , Actinas/antagonistas & inibidores , Actinas/genética , Actinas/metabolismo , Animais , Animais Geneticamente Modificados , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/ultraestrutura , Encéfalo/irrigação sanguínea , Encéfalo/embriologia , Encéfalo/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Circulação Cerebrovascular/genética , Embrião não Mamífero , Células Endoteliais/metabolismo , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Miosina Tipo II/antagonistas & inibidores , Miosina Tipo II/genética , Miosina Tipo II/metabolismo , Óxido Nítrico/metabolismo , Organelas/metabolismo , Organelas/ultraestrutura , Polimerização/efeitos dos fármacos , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais , Tiazolidinas/farmacologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
14.
Dev Cell ; 32(2): 181-90, 2015 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-25625207

RESUMO

Mechanotransduction pathways are activated in response to biophysical stimuli during the development or homeostasis of organs and tissues. In zebrafish, the blood-flow-sensitive transcription factor Klf2a promotes VEGF-dependent angiogenesis. However, the means by which the Klf2a mechanotransduction pathway is regulated to prevent continuous angiogenesis remain unknown. Here we report that the upregulation of klf2 mRNA causes enhanced egfl7 expression and angiogenesis signaling, which underlies cardiovascular defects associated with the loss of cerebral cavernous malformation (CCM) proteins in the zebrafish embryo. Using CCM-protein-depleted human umbilical vein endothelial cells, we show that the misexpression of KLF2 mRNA requires the extracellular matrix-binding receptor ß1 integrin and occurs in the absence of blood flow. Downregulation of ß1 integrin rescues ccm mutant cardiovascular malformations in zebrafish. Our work reveals a ß1 integrin-Klf2-Egfl7-signaling pathway that is tightly regulated by CCM proteins. This regulation prevents angiogenic overgrowth and ensures the quiescence of endothelial cells.


Assuntos
Movimento Celular/fisiologia , Hemangioma Cavernoso do Sistema Nervoso Central/metabolismo , Integrina beta1/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Neovascularização Patológica/metabolismo , Proteínas/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Ligação ao Cálcio , Adesão Celular/fisiologia , Movimento Celular/genética , Neoplasias do Sistema Nervoso Central/metabolismo , Família de Proteínas EGF , Hemangioma Cavernoso do Sistema Nervoso Central/genética , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Mecanotransdução Celular/fisiologia , Proteínas de Membrana/metabolismo , Camundongos , Proteínas do Tecido Nervoso/metabolismo , RNA Interferente Pequeno/genética , Transdução de Sinais/fisiologia , Peixe-Zebra
15.
Development ; 137(22): 3867-75, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20978078

RESUMO

Cardiac trabeculation is a crucial morphogenetic process by which clusters of ventricular cardiomyocytes extrude and expand into the cardiac jelly to form sheet-like projections. Although it has been suggested that cardiac trabeculae enhance cardiac contractility and intra-ventricular conduction, their exact function in heart development has not been directly addressed. We found that in zebrafish erbb2 mutants, which we show completely lack cardiac trabeculae, cardiac function is significantly compromised, with mutant hearts exhibiting decreased fractional shortening and an immature conduction pattern. To begin to elucidate the cellular mechanisms of ErbB2 function in cardiac trabeculation, we analyzed erbb2 mutant hearts more closely and found that loss of ErbB2 activity resulted in a complete absence of cardiomyocyte proliferation during trabeculation stages. In addition, based on data obtained from proliferation, lineage tracing and transplantation studies, we propose that cardiac trabeculation is initiated by directional cardiomyocyte migration rather than oriented cell division, and that ErbB2 cell-autonomously regulates this process.


Assuntos
Coração/embriologia , Morfogênese , Miócitos Cardíacos/metabolismo , Receptor ErbB-2/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Animais , Movimento Celular , Proliferação de Células , Miocárdio/metabolismo
16.
Curr Biol ; 20(20): 1840-5, 2010 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-20933420

RESUMO

Transport of chloride through the cystic fibrosis transmembrane conductance regulator (CFTR) channel is a key step in regulating fluid secretion in vertebrates [1, 2]. Loss of CFTR function leads to cystic fibrosis [1, 3, 4], a disease that affects the lungs, pancreas, liver, intestine, and vas deferens. Conversely, uncontrolled activation of the channel leads to increased fluid secretion and plays a major role in several diseases and conditions including cholera [5, 6] and other secretory diarrheas [7] as well as polycystic kidney disease [8-10]. Understanding how CFTR activity is regulated in vivo has been limited by the lack of a genetic model. Here, we used a forward genetic approach in zebrafish to uncover CFTR regulators. We report the identification, isolation, and characterization of a mutation in the zebrafish cse1l gene that leads to the sudden and dramatic expansion of the gut tube. We show that this phenotype results from a rapid accumulation of fluid due to the uncontrolled activation of the CFTR channel. Analyses in zebrafish larvae and mammalian cells indicate that Cse1l is a negative regulator of CFTR-dependent fluid secretion. This work demonstrates the importance of fluid homeostasis in development and establishes the zebrafish as a much-needed model system to study CFTR regulation in vivo.


Assuntos
Líquidos Corporais/metabolismo , Proteína de Suscetibilidade a Apoptose Celular/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Trato Gastrointestinal/metabolismo , Homeostase/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Animais , Linhagem Celular , Cães , Trato Gastrointestinal/anormalidades , Trato Gastrointestinal/embriologia , Genes Recessivos , Proteínas de Fluorescência Verde , Imunoprecipitação , Microscopia Confocal , Mutação/genética , Peixe-Zebra
17.
Science ; 326(5950): 294-8, 2009 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-19815777

RESUMO

Blood vessels form de novo (vasculogenesis) or upon sprouting of capillaries from preexisting vessels (angiogenesis). With high-resolution imaging of zebrafish vascular development, we uncovered a third mode of blood vessel formation whereby the first embryonic artery and vein, two unconnected blood vessels, arise from a common precursor vessel. The first embryonic vein formed by selective sprouting of progenitor cells from the precursor vessel, followed by vessel segregation. These processes were regulated by the ligand EphrinB2 and its receptor EphB4, which are expressed in arterial-fated and venous-fated progenitors, respectively, and interact to orient the direction of progenitor migration. Thus, directional control of progenitor migration drives arterial-venous segregation and generation of separate parallel vessels from a single precursor vessel, a process essential for vascular development.


Assuntos
Artérias/embriologia , Células Endoteliais/fisiologia , Efrina-B2/metabolismo , Morfogênese , Receptor EphB4/metabolismo , Células-Tronco/fisiologia , Veias/embriologia , Animais , Animais Geneticamente Modificados , Aorta/citologia , Aorta/embriologia , Artérias/citologia , Movimento Celular , Células Endoteliais/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Veias/citologia , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
18.
Circ Res ; 104(11): 1260-6, 2009 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-19423847

RESUMO

In a recent genetic screen, we identified mutations in genes important for vascular development and maintenance in zebrafish (Jin et al. Dev Biol. 2007;307:29-42). Mutations [corrected] at the adrasteia (adr) locus cause a pronounced dilatation of the aortic arch vessels as well as aberrant patterning of the hindbrain capillaries and, to a lesser extent, intersomitic vessels. This dilatation of the aortic arch vessels does not appear to be caused by increased cell proliferation but is dependent on vascular endothelial growth factor (Vegf) signaling. By positional cloning, we isolated seryl-tRNA synthetase (sars) as the gene affected by the adr mutations. Small interfering RNA knockdown experiments in human umbilical vein endothelial cell cultures indicate that SARS also regulates endothelial sprouting. These analyses of zebrafish and human endothelial cells reveal a new noncanonical function of Sars in endothelial development.


Assuntos
Circulação Cerebrovascular/fisiologia , Coração/embriologia , Serina-tRNA Ligase/genética , Inibidores da Angiogênese/farmacologia , Animais , Aorta Torácica/citologia , Aorta Torácica/embriologia , Aorta Torácica/fisiologia , Encéfalo/fisiologia , Divisão Celular , Mapeamento Cromossômico , Cicloeximida/farmacologia , Embrião não Mamífero/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Humanos , Indóis/farmacologia , Miocárdio/citologia , Pirróis/farmacologia , RNA Interferente Pequeno/genética , Transfecção , Veias Umbilicais/citologia , Veias Umbilicais/fisiologia , Peixe-Zebra
19.
Curr Biol ; 19(12): 1034-9, 2009 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-19464178

RESUMO

Members of the Hedgehog (Hh) family of secreted proteins function as morphogens to pattern developing tissues and control cell proliferation. The seven-transmembrane domain (7TM) protein Smoothened (Smo) is essential for the activation of all levels of Hh signaling. However, the mechanisms by which Smo differentially activates low- or high-level Hh signaling are not known. Here we show that a newly identified mutation in the extracellular domain (ECD) of zebrafish Smo attenuates Smo signaling. The Smo agonist purmorphamine induces the stabilization, ciliary translocation, and high-level signaling of wild-type Smo. In contrast, purmorphamine induces the stabilization but not the ciliary translocation or high-level signaling of the Smo ECD mutant protein. Surprisingly, a truncated form of Smo that lacks the cysteine-rich domain of the ECD localizes to the cilium but is unable to activate high-level Hh signaling. We also present evidence that cilia may be required for Hh signaling in early zebrafish embryos. These data indicate that the ECD, previously thought to be dispensable for vertebrate Smo function, both regulates Smo ciliary localization and is essential for high-level Hh signaling.


Assuntos
Cílios , Proteínas Hedgehog/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Sequência de Aminoácidos , Animais , Cílios/fisiologia , Cílios/ultraestrutura , Proteínas Hedgehog/genética , Dados de Sequência Molecular , Morfolinas/metabolismo , Estrutura Terciária de Proteína , Purinas/metabolismo , Receptores de Superfície Celular/genética , Receptores Acoplados a Proteínas G/genética , Receptor Smoothened , Peixe-Zebra/anatomia & histologia , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
20.
Proc Natl Acad Sci U S A ; 104(27): 11316-21, 2007 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-17592134

RESUMO

Long QT syndrome (LQTS) is a disorder of ventricular repolarization that predisposes affected individuals to lethal cardiac arrhythmias. To date, an appropriate animal model of inherited LQTS does not exist. The zebrafish is a powerful vertebrate model used to dissect molecular pathways of cardiovascular development and disease. Because fundamental electrical properties of the zebrafish heart are remarkably similar to those of the human heart, the zebrafish may be an appropriate model for studying human inherited arrhythmias. Here we describe the molecular, cellular, and electrophysiological basis of a zebrafish mutant characterized by ventricular asystole. Genetic mapping and direct sequencing identify the affected gene as kcnh2, which encodes the channel responsible for the rapidly activating delayed rectifier K(+) current (I(Kr)). We show that complete loss of functional I(Kr) in embryonic hearts leads to ventricular cell membrane depolarization, inability to generate action potentials (APs), and disrupted calcium release. A small hyperpolarizing current restores spontaneous APs, implying wild-type function of other ionic currents critical for AP generation. Heterozygous fish manifest overt cellular and electrocardiographic evidence for delayed ventricular repolarization. Our findings provide insight into the pathogenesis of homozygous kcnh2 mutations and expand the use of zebrafish mutants as a model system to study human arrhythmias.


Assuntos
Modelos Animais de Doenças , Síndrome do QT Longo/genética , Peixe-Zebra/genética , Potenciais de Ação/genética , Substituição de Aminoácidos/genética , Animais , Canal de Potássio ERG1 , Eletrocardiografia , Canais de Potássio Éter-A-Go-Go , Parada Cardíaca/genética , Parada Cardíaca/fisiopatologia , Humanos , Síndrome do QT Longo/embriologia , Síndrome do QT Longo/fisiopatologia , Fenótipo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Xenopus , Peixe-Zebra/embriologia , Peixe-Zebra/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA