Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Dis Model Mech ; 16(3)2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36805642

RESUMO

People with activated PI3 kinase delta syndrome 1 (APDS1) suffer from immune deficiency and severe bronchiectasis. APDS1 is caused by dominant activating mutations of the PIK3CD gene that encodes the PI3 kinase delta (PI3Kδ) catalytic subunit. Despite the importance of innate immunity defects in bronchiectasis, there has been limited investigation of neutrophils or macrophages in APDS1 patients or mouse models. Zebrafish embryos provide an ideal system to study neutrophils and macrophages. We used CRISPR-Cas9 and CRISPR-Cpf1, with oligonucleotide-directed homologous repair, to engineer zebrafish equivalents of the two most prevalent human APDS1 disease mutations. These zebrafish pik3cd alleles dominantly caused excessive neutrophilic inflammation in a tail-fin injury model. They also resulted in total body neutrophilia in the absence of any inflammatory stimulus but normal numbers of macrophages. Exposure of zebrafish to the PI3Kδ inhibitor CAL-101 reversed the total body neutrophilia. There was no apparent defect in neutrophil maturation or migration, and tail-fin regeneration was unimpaired. Overall, the finding is of enhanced granulopoeisis, in the absence of notable phenotypic change in neutrophils and macrophages.


Assuntos
Bronquiectasia , Peixe-Zebra , Animais , Camundongos , Humanos , Peixe-Zebra/genética , Fosfatidilinositol 3-Quinases , Mutação , Neutrófilos
2.
Front Cell Dev Biol ; 9: 676214, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34268305

RESUMO

Cilia are evolutionarily highly conserved organelles with important functions in many organs. The extracellular component of the cilium protruding from the plasma membrane comprises an axoneme composed of microtubule doublets, arranged in a 9 + 0 conformation in primary cilia or 9 + 2 in motile cilia. These microtubules facilitate transport of intraflagellar cargoes along the axoneme. They also provide structural stability to the cilium, which may play an important role in sensory cilia, where signals are received from the movement of extracellular fluid. Post-translational modification of microtubules in cilia is a well-studied phenomenon, and acetylation on lysine 40 (K40) of alpha tubulin is prominent in cilia. It is believed that this modification contributes to the stabilization of cilia. Two classes of enzymes, histone acetyltransferases and histone deacetylases, mediate regulation of tubulin acetylation. Here we use a genetic approach, immunocytochemistry and behavioral tests to investigate the function of tubulin deacetylases in cilia in a zebrafish model. By mutating three histone deacetylase genes (Sirt2, Hdac6, and Hdac10), we identify an unforeseen role for Hdac6 and Sirt2 in cilia. As expected, mutation of these genes leads to increased acetylation of cytoplasmic tubulin, however, surprisingly it caused decreased tubulin acetylation in cilia in the developing eye, ear, brain and kidney. Cilia in the ear and eye showed elevated levels of mono-glycylated tubulin suggesting a compensatory mechanism. These changes did not affect the length or morphology of cilia, however, functional defects in balance was observed, suggesting that the level of tubulin acetylation may affect function of the cilium.

3.
Dev Dyn ; 248(6): 410-425, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30980582

RESUMO

BACKGROUND: Cilia are essential for morphogenesis and maintenance of many tissues. Loss-of-function of cilia in early Zebrafish development causes a range of vascular defects, including cerebral hemorrhage and reduced arterial vascular mural cell coverage. In contrast, loss of endothelial cilia in mice has little effect on vascular development. We therefore used a conditional rescue approach to induce endothelial cilia ablation after early embryonic development and examined the effect on vascular development and mural cell development in postembryonic, juvenile, and adult Zebrafish. RESULTS: ift54(elipsa)-mutant Zebrafish are unable to form cilia. We rescued cilia formation and ameliorated the phenotype of ift54 mutants using a novel Tg(ubi:loxP-ift54-loxP-myr-mcherry,myl7:EGFP)sh488 transgene expressing wild-type ift54 flanked by recombinase sites, then used a Tg(kdrl:cre)s898 transgene to induce endothelial-specific inactivation of ift54 at postembryonic ages. Fish without endothelial ift54 function could survive to adulthood and exhibited no vascular defects. Endothelial inactivation of ift54 did not affect development of tagln-positive vascular mural cells around either the aorta or the caudal fin vessels, or formation of vessels after tail fin resection in adult animals. CONCLUSIONS: Endothelial cilia are not essential for development and remodeling of the vasculature in juvenile and adult Zebrafish when inactivated after embryogenesis.


Assuntos
Endotélio Vascular , Animais
4.
Blood Adv ; 2(20): 2589-2606, 2018 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-30309860

RESUMO

The transcriptional repressors Gfi1(a) and Gfi1b are epigenetic regulators with unique and overlapping roles in hematopoiesis. In different contexts, Gfi1 and Gfi1b restrict or promote cell proliferation, prevent apoptosis, influence cell fate decisions, and are essential for terminal differentiation. Here, we show in primitive red blood cells (prRBCs) that they can also set the pace for cellular differentiation. In zebrafish, prRBCs express 2 of 3 zebrafish Gfi1/1b paralogs, Gfi1aa and Gfi1b. The recently identified zebrafish gfi1aa gene trap allele qmc551 drives erythroid green fluorescent protein (GFP) instead of Gfi1aa expression, yet homozygous carriers have normal prRBCs. prRBCs display a maturation defect only after splice morpholino-mediated knockdown of Gfi1b in gfi1aa qmc551 homozygous embryos. To study the transcriptome of the Gfi1aa/1b double-depleted cells, we performed an RNA-Seq experiment on GFP-positive prRBCs sorted from 20-hour-old embryos that were heterozygous or homozygous for gfi1aa qmc551 , as well as wt or morphant for gfi1b We subsequently confirmed and extended these data in whole-mount in situ hybridization experiments on newly generated single- and double-mutant embryos. Combined, the data showed that in the absence of Gfi1aa, the synchronously developing prRBCs were delayed in activating late erythroid differentiation, as they struggled to suppress early erythroid and endothelial transcription programs. The latter highlighted the bipotent nature of the progenitors from which prRBCs arise. In the absence of Gfi1aa, Gfi1b promoted erythroid differentiation as stepwise loss of wt gfi1b copies progressively delayed Gfi1aa-depleted prRBCs even further, showing that Gfi1aa and Gfi1b together set the pace for prRBC differentiation from hemangioblasts.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos/metabolismo , Eritroblastos/metabolismo , Hemangioblastos/metabolismo , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular , Peixe-Zebra
5.
PLoS One ; 10(10): e0141611, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26506092

RESUMO

INTRODUCTION AND OBJECTIVES: The zinc-finger transcription factor Krϋppel-like factor 2 (KLF2) transduces blood flow into molecular signals responsible for a wide range of responses within the vasculature. KLF2 maintains a healthy, quiescent endothelial phenotype. Previous studies report a range of phenotypes following morpholino antisense oligonucleotide-induced klf2a knockdown in zebrafish. Targeted genome editing is an increasingly applied method for functional assessment of candidate genes. We therefore generated a stable klf2a mutant zebrafish and characterised its cardiovascular and haematopoietic development. METHODS AND RESULTS: Using Transcription Activator-Like Effector Nucleases (TALEN) we generated a klf2a mutant (klf2ash317) with a 14bp deletion leading to a premature stop codon in exon 2. Western blotting confirmed loss of wild type Klf2a protein and the presence of a truncated protein in klf2ash317 mutants. Homozygous klf2ash317 mutants exhibit no defects in vascular patterning, survive to adulthood and are fertile, without displaying previously described morphant phenotypes such as high-output cardiac failure, reduced haematopoetic stem cell (HSC) development or impaired formation of the 5th accessory aortic arch. Homozygous klf2ash317 mutation did not reduce angiogenesis in zebrafish with homozygous mutations in von Hippel Lindau (vhl), a form of angiogenesis that is dependent on blood flow. We examined expression of three klf family members in wildtype and klf2ash317 zebrafish. We detected vascular expression of klf2b (but not klf4a or biklf/klf4b/klf17) in wildtypes but found no differences in expression that might account for the lack of phenotype in klf2ash317 mutants. klf2b morpholino knockdown did not affect heart rate or impair formation of the 5th accessory aortic arch in either wildtypes or klf2ash317 mutants. CONCLUSIONS: The klf2ash317 mutation produces a truncated Klf2a protein but, unlike morpholino induced klf2a knockdown, does not affect cardiovascular development.


Assuntos
Sistema Cardiovascular/crescimento & desenvolvimento , Sistema Hematopoético/crescimento & desenvolvimento , Fatores de Transcrição Kruppel-Like/genética , Morfogênese/genética , Proteínas de Peixe-Zebra/genética , Animais , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Humanos , Fatores de Transcrição Kruppel-Like/antagonistas & inibidores , Fatores de Transcrição Kruppel-Like/biossíntese , Morfolinos/genética , Mutação , Transdução de Sinais , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/biossíntese
6.
Skelet Muscle ; 5(1): 2, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25671076

RESUMO

BACKGROUND: The transcription factor Sox6 has been implicated in regulating muscle fiber type-specific gene expression in mammals. In zebrafish, loss of function of the transcription factor Prdm1a results in a slow to fast-twitch fiber type transformation presaged by ectopic expression of sox6 in slow-twitch progenitors. Morpholino-mediated Sox6 knockdown can suppress this transformation but causes ectopic expression of only one of three slow-twitch specific genes assayed. Here, we use gain and loss of function analysis to analyse further the role of Sox6 in zebrafish muscle fiber type specification. METHODS: The GAL4 binary misexpression system was used to express Sox6 ectopically in zebrafish embryos. Cis-regulatory elements were characterized using transgenic fish. Zinc finger nuclease mediated targeted mutagenesis was used to analyse the effects of loss of Sox6 function in embryonic, larval and adult zebrafish. Zebrafish transgenic for the GCaMP3 Calcium reporter were used to assay Ca2+ transients in wild-type and mutant muscle fibres. RESULTS: Ectopic Sox6 expression is sufficient to downregulate slow-twitch specific gene expression in zebrafish embryos. Cis-regulatory elements upstream of the slow myosin heavy chain 1 (smyhc1) and slow troponin c (tnnc1b) genes contain putative Sox6 binding sites required for repression of the former but not the latter. Embryos homozygous for sox6 null alleles expressed tnnc1b throughout the fast-twitch muscle whereas other slow-specific muscle genes, including smyhc1, were expressed ectopically in only a subset of fast-twitch fibers. Ca2+ transients in sox6 mutant fast-twitch fibers were intermediate in their speed and amplitude between those of wild-type slow- and fast-twitch fibers. sox6 homozygotes survived to adulthood and exhibited continued misexpression of tnnc1b as well as smaller slow-twitch fibers. They also exhibited a striking curvature of the spine. CONCLUSIONS: The Sox6 transcription factor is a key regulator of fast-twitch muscle fiber differentiation in the zebrafish, a role similar to that ascribed to its murine ortholog.

7.
PLoS Genet ; 9(12): e1003955, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24339784

RESUMO

Loss of function mutations of Kif7, the vertebrate orthologue of the Drosophila Hh pathway component Costal2, cause defects in the limbs and neural tubes of mice, attributable to ectopic expression of Hh target genes. While this implies a functional conservation of Cos2 and Kif7 between flies and vertebrates, the association of Kif7 with the primary cilium, an organelle absent from most Drosophila cells, suggests their mechanisms of action may have diverged. Here, using mutant alleles induced by Zinc Finger Nuclease-mediated targeted mutagenesis, we show that in zebrafish, Kif7 acts principally to suppress the activity of the Gli1 transcription factor. Notably, we find that endogenous Kif7 protein accumulates not only in the primary cilium, as previously observed in mammalian cells, but also in cytoplasmic puncta that disperse in response to Hh pathway activation. Moreover, we show that Drosophila Costal2 can substitute for Kif7, suggesting a conserved mode of action of the two proteins. We show that Kif7 interacts with both Gli1 and Gli2a and suggest that it functions to sequester Gli proteins in the cytoplasm, in a manner analogous to the regulation of Ci by Cos2 in Drosophila. We also show that zebrafish Kif7 potentiates Gli2a activity by promoting its dissociation from the Suppressor of Fused (Sufu) protein and present evidence that it mediates a Smo dependent modification of the full length form of Gli2a. Surprisingly, the function of Kif7 in the zebrafish embryo appears restricted principally to mesodermal derivatives, its inactivation having little effect on neural tube patterning, even when Sufu protein levels are depleted. Remarkably, zebrafish lacking all Kif7 function are viable, in contrast to the peri-natal lethality of mouse kif7 mutants but similar to some Acrocallosal or Joubert syndrome patients who are homozygous for loss of function KIF7 alleles.


Assuntos
Cílios/genética , Cinesinas/genética , Proteínas Oncogênicas/genética , Transativadores/genética , Fatores de Transcrição/genética , Proteínas de Peixe-Zebra/genética , Anormalidades Múltiplas , Animais , Doenças Cerebelares/genética , Doenças Cerebelares/patologia , Cerebelo/anormalidades , Embrião não Mamífero/metabolismo , Extremidades/crescimento & desenvolvimento , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Doenças Renais Císticas/genética , Doenças Renais Císticas/patologia , Cinesinas/metabolismo , Camundongos , Tubo Neural/crescimento & desenvolvimento , Proteínas Oncogênicas/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Retina/anormalidades , Retina/patologia , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Proteínas de Peixe-Zebra/metabolismo , Proteína GLI1 em Dedos de Zinco , Proteína Gli2 com Dedos de Zinco
8.
Biotechniques ; 55(6): 314-6, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24344681

RESUMO

Here we describe a method for high-throughput genotyping of live larval zebrafish as early as 72 h post-fertilization (hpf). Importantly, this technique allows rapid and cost-effective PCR-based genotyping from very small fin biopsies, which regenerate as the embryo develops, thereby allowing researchers to select embryos with desired genotypes to be raised to adulthood.


Assuntos
Técnicas de Genotipagem , Peixe-Zebra/genética , Animais , Biópsia/veterinária , Sequenciamento de Nucleotídeos em Larga Escala , Larva/genética , Reação em Cadeia da Polimerase/métodos , Cauda/patologia , Peixe-Zebra/crescimento & desenvolvimento
9.
Cancer Res ; 72(16): 4017-27, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22665266

RESUMO

Hypoxic signaling is a central modulator of cellular physiology in cancer. Core members of oxygen-sensing pathway including the von Hippel-Lindau tumor suppressor protein (pVHL) and the hypoxia inducible factor (HIF) transcription factors have been intensively studied, but improved organismal models might speed advances for both pathobiologic understanding and therapeutic modulation. To study HIF signaling during tumorigenesis and development in zebrafish, we developed a unique in vivo reporter for hypoxia, expressing EGFP driven by prolyl hydroxylase 3 (phd3) promoter/regulatory elements. Modulation of HIF pathway in Tg(phd3::EGFP) embryos showed a specific role for hypoxic signaling in the transgene activation. Zebrafish vhl mutants display a systemic hypoxia response, reflected by strong and ubiquitous transgene expression. In contrast to human VHL patients, heterozygous Vhl mice and vhl zebrafish are not predisposed to cancer. However, upon exposure to dimethylbenzanthracene (DMBA), the vhl heterozygous fish showed an increase in the occurrence of hepatic and intestinal tumors, a subset of which exhibited strong transgene expression, suggesting loss of Vhl function in these tumor cells. Compared with control fish, DMBA-treated vhl heterozygous fish also showed an increase in proliferating cell nuclear antigen-positive renal tubules. Taken together, our findings establish Vhl as a genuine tumor suppressor in zebrafish and offer this model as a tool to noninvasively study VHL and HIF signaling during tumorigenesis and development.


Assuntos
Transformação Celular Neoplásica/metabolismo , Modelos Animais de Doenças , Neoplasias Intestinais/metabolismo , Neoplasias Hepáticas Experimentais/metabolismo , 9,10-Dimetil-1,2-benzantraceno , Animais , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , Transformação Celular Neoplásica/induzido quimicamente , Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Intestinais/induzido quimicamente , Neoplasias Hepáticas Experimentais/induzido quimicamente , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo , Proteínas de Peixe-Zebra/metabolismo
10.
Development ; 138(22): 4969-78, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22028029

RESUMO

Using zinc-finger nuclease-mediated mutagenesis, we have generated mutant alleles of the zebrafish orthologue of the chicken talpid3 (ta3) gene, which encodes a centrosomal protein that is essential for ciliogenesis. Animals homozygous for these mutant alleles complete embryogenesis normally, but manifest a cystic kidney phenotype during the early larval stages and die within a month of hatching. Elimination of maternally derived Ta3 activity by germline replacement resulted in embryonic lethality of ta3 homozygotes. The phenotype of such maternal and zygotic (MZta3) mutant zebrafish showed strong similarities to that of chick ta3 mutants: absence of primary and motile cilia as well as aberrant Hedgehog (Hh) signalling, the latter manifest by the expanded domains of engrailed and ptc1 expression in the somites, reduction of nkx2.2 expression in the neural tube, symmetric pectoral fins, cyclopic eyes and an ectopic lens. GFP-tagged Gli2a localised to the basal bodies in the absence of the primary cilia and western blot analysis showed that Gli2a protein is aberrantly processed in MZta3 embryos. Zygotic expression of ta3 largely rescued the effects of maternal depletion, but the motile cilia of Kupffer's vesicle remained aberrant, resulting in laterality defects. Our findings underline the importance of the primary cilium for Hh signaling in zebrafish and reveal the conservation of Ta3 function during vertebrate evolution.


Assuntos
Cílios/genética , Proteínas Hedgehog/genética , Vertebrados/genética , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/genética , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Sequência de Bases , Galinhas/genética , Cílios/fisiologia , Sequência Conservada/fisiologia , Embrião não Mamífero , Feminino , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/fisiologia , Proteína Homeobox Nkx-2.2 , Masculino , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Organogênese/genética , Homologia de Sequência de Aminoácidos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Vertebrados/embriologia , Peixe-Zebra/embriologia , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra/genética
11.
Dev Dyn ; 240(11): 2440-51, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21954137

RESUMO

The transcription factor Pax7 is a marker and regulator of muscle progenitors and satellite cells that contribute to the embryonic development and postembryonic growth of skeletal muscle in vertebrates, as well as to its repair and regeneration. Here, we identify Pax7(+ve) myogenic cells in the zebrafish and characterize their behavior in postembryonic stages. Mononucleate Pax7(+ve) cells can first be found associated with myofibers at 72 hours post fertilization (hpf). To follow the behavior of muscle progenitor cells in vivo, we generated transgenic lines expressing fluorescent proteins under the control of the pax7a or pax3a promoters. We established an injury model using cardiotoxin injection and monitored cell proliferation and myogenic regulatory factor expression in myogenic precursors cells and muscle fibers after injury using proliferation markers and the transgenic lines. We also analyzed Pax7(+ve) cells in animals with dystrophic phenotypes and found an increased number compared with wild-type.


Assuntos
Modelos Animais de Doenças , Desenvolvimento Muscular/genética , Doenças Musculares/genética , Mioblastos/metabolismo , Fator de Transcrição PAX7/genética , Peixe-Zebra/embriologia , Animais , Animais Geneticamente Modificados , Proteínas Cardiotóxicas de Elapídeos , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter/fisiologia , Humanos , Músculo Esquelético/lesões , Músculo Esquelético/metabolismo , Doenças Musculares/induzido quimicamente , Doenças Musculares/metabolismo , Doenças Musculares/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia , Mioblastos/fisiologia , Fator de Transcrição PAX7/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
12.
EMBO Rep ; 9(7): 683-9, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18535625

RESUMO

The zebrafish u-boot (ubo) gene encodes the transcription factor Prdm1, which is essential for the specification of the primary slow-twitch muscle fibres that derive from adaxial cells. Here, we show that Prdm1 functions by acting as a transcriptional repressor and that slow-twitch-specific muscle gene expression is activated by Prdm1-mediated repression of the transcriptional repressor Sox6. Genes encoding fast-specific isoforms of sarcomeric proteins are ectopically expressed in the adaxial cells of ubo(tp39) mutant embryos. By using chromatin immunoprecipitation, we show that these are direct targets of Prdm1. Thus, Prdm1 promotes slow-twitch fibre differentiation by acting as a global repressor of fast-fibre-specific genes, as well as by abrogating the repression of slow-fibre-specific genes.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Embrião não Mamífero/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Transcrição Gênica , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Sítios de Ligação , Miosinas Cardíacas/genética , Diferenciação Celular , Imunoprecipitação da Cromatina , Regulação da Expressão Gênica no Desenvolvimento , Fibras Musculares de Contração Rápida/citologia , Fibras Musculares Esqueléticas/citologia , Fibras Musculares de Contração Lenta/citologia , Mutação/genética , Cadeias Leves de Miosina/genética , Especificidade de Órgãos , Fator 1 de Ligação ao Domínio I Regulador Positivo , Regiões Promotoras Genéticas/genética , Regulação para Cima/genética , Peixe-Zebra/genética
13.
Development ; 135(12): 2115-26, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18480160

RESUMO

The zebrafish embryo develops a series of anatomically distinct slow twitch muscle fibres that characteristically express genes encoding lineage-specific isoforms of sarcomeric proteins such as MyHC and troponin. We show here that different subsets of these slow fibres express distinct members of a tandem array of slow MyHC genes. The first slow twitch muscle fibres to differentiate, which are specified by the activity of the transcription factor Prdm1 (also called Ubo or Blimp1) in response to Hedgehog (Hh) signalling, express the smyhc1 gene. Subsequently, secondary slow twitch fibres differentiate in most cases independently of Hh activity. We find that although some of these later-forming fibres also express smyhc1, others express smyhc2 or smyhc3. We show that the smyhc1-positive fibres express the ubo (prdm1) gene and adopt fast twitch fibre characteristics in the absence of Prdm1 activity, whereas those that do not express smyhc1 can differentiate independently of Prdm1 function. Conversely, some smyhc2-expressing fibres, although independent of Prdm1 function, require Hh activity to form. The adult trunk slow fibres express smyhc2 and smyhc3, but lack smyhc1 expression. The different slow fibres in the craniofacial muscles variously express smyhc1, smyhc2 and smyhc3, and all differentiate independently of Prdm1.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Proteínas Hedgehog/fisiologia , Fibras Musculares de Contração Lenta/fisiologia , Cadeias Pesadas de Miosina/genética , Proteínas Nucleares/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/embriologia , Animais , Proteínas de Ligação a DNA/genética , Embrião não Mamífero , Técnica Direta de Fluorescência para Anticorpo , Expressão Gênica/fisiologia , Heterogeneidade Genética , Proteínas Hedgehog/genética , Hibridização In Situ , Fibras Musculares de Contração Lenta/citologia , Cadeias Pesadas de Miosina/metabolismo , Proteínas Nucleares/genética , Fator 1 de Ligação ao Domínio I Regulador Positivo , Transgenes , Proteínas de Peixe-Zebra/genética
14.
Exp Lung Res ; 33(10): 549-54, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18075830

RESUMO

Neutrophilic inflammation is responsible for much of the tissue damage seen in many lung diseases. For resolution of inflammation to occur, neutrophils must die by apoptosis, allowing their recognition and removal by macrophages. The molecular events controlling this important regulatory step are poorly understood, in large part due to the genetic intractability of the human neutrophil granulocyte. The authors have established a model of inflammation in the Zebrafish, which shares many features of the innate immune system with those of humans. Injury to the Zebrafish tailfin induces a reproducible and quantifiable inflammatory response, which resolves with kinetics similar to mammalian models of neutrophilic inflammation, including pulmonary inflammation. Pharmacological modulation of neutrophil apoptosis can modulate the outcome of experimentally induced inflammation. In addition, the authors have generated a construct that expresses green fluorescent protein under the myeloperoxidase promoter, allowing in vivo visualization of neutrophils during experimentally induced inflammation. The authors are also performing an unbiased forward genetic screen for mutants with defective resolution of inflammation, and to date have identified a number of putative mutants. Further study and characterization of these mutants is underway. The authors have thus established an important experimental link between apoptosis and resolution of inflammation in an in vivo system, and defined an important new model for the study of inflammation resolution. The authors hope that these tools will permit detailed study of the genetic controls of the resolution of inflammation, and provide insights with potential clinical utility.


Assuntos
Modelos Animais de Doenças , Inflamação/imunologia , Pneumopatias/imunologia , Peixe-Zebra/imunologia , Animais , Apoptose/fisiologia , Inflamação/patologia , Inflamação/fisiopatologia , Pneumopatias/patologia , Pneumopatias/fisiopatologia , Neutrófilos/patologia
15.
Development ; 134(5): 1011-22, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17267445

RESUMO

Pigment pattern formation in zebrafish presents a tractable model system for studying the morphogenesis of neural crest derivatives. Embryos mutant for choker manifest a unique pigment pattern phenotype that combines a loss of lateral stripe melanophores with an ectopic melanophore ;collar' at the head-trunk border. We find that defects in neural crest migration are largely restricted to the lateral migration pathway, affecting both xanthophores (lost) and melanophores (gained) in choker mutants. Double mutant and timelapse analyses demonstrate that these defects are likely to be driven independently, the collar being formed by invasion of melanophores from the dorsal and ventral stripes. Using tissue transplantation, we show that melanophore patterning depends upon the underlying somitic cells, the myotomal derivatives of which--both slow--and fast-twitch muscle fibres--are themselves significantly disorganised in the region of the ectopic collar. In addition, we uncover an aberrant pattern of expression of the gene encoding the chemokine Sdf1a in choker mutant homozygotes that correlates with each aspect of the melanophore pattern defect. Using morpholino knock-down and ectopic expression experiments, we provide evidence to suggest that Sdf1a drives melanophore invasion in the choker mutant collar and normally plays an essential role in patterning the lateral stripe. We thus identify Sdf1 as a key molecule in pigment pattern formation, adding to the growing inventory of its roles in embryonic development.


Assuntos
Quimiocinas CXC/fisiologia , Melanóforos/fisiologia , Crista Neural/fisiologia , Somitos/fisiologia , Peixe-Zebra/fisiologia , Animais , Padronização Corporal , Movimento Celular , Quimiocina CXCL12 , Quimiocinas CXC/genética , Quimiocinas CXC/metabolismo , Melanóforos/citologia , Fibras Musculares de Contração Rápida/fisiologia , Fibras Musculares de Contração Lenta/fisiologia , Mutação , Pigmentação , Somitos/citologia , Peixe-Zebra/embriologia , Peixe-Zebra/genética
16.
Blood ; 108(13): 3976-8, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16926288

RESUMO

We have established an in vivo model for genetic analysis of the inflammatory response by generating a transgenic zebrafish line that expresses GFP under the neutrophil-specific myeloperoxidase promoter. We show that inflammation is induced after transection of the tail of zebrafish larvae and that this inflammation subsequently resolves over a similar time course to mammalian systems. Quantitative data can be generated from this model by counting of fluorescent cells or by digital image analysis. In addition, we show that the resolution of experimentally induced inflammation can be inhibited by the addition of a pancaspase inhibitor, zVD.fmk, demonstrating that experimental manipulation of the resolution of inflammation is possible in this model.


Assuntos
Neutrófilos/imunologia , Ferimentos e Lesões/imunologia , Peixe-Zebra/imunologia , Animais , Animais Geneticamente Modificados , Contagem de Células/métodos , Modelos Animais de Doenças , Proteínas de Fluorescência Verde , Processamento de Imagem Assistida por Computador , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Neutrófilos/patologia , Peroxidase/genética , Peroxidase/imunologia , Regiões Promotoras Genéticas , Transgenes/imunologia , Ferimentos e Lesões/genética , Ferimentos e Lesões/patologia , Peixe-Zebra/genética
17.
Mech Dev ; 122(5): 659-69, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15817223

RESUMO

Zebrafish lacking functional sox10 have defects in non-ectomesenchymal neural crest derivatives including the enteric nervous system (ENS) and as such provide an animal model for human Waardenburg Syndrome IV. Here, we characterize zebrafish phox2b as a functionally conserved marker of the developing ENS. We show that morpholino-mediated knockdown of Phox2b generates fish modeling Hirschsprung disease. Using markers, including phox2b, we investigate the ontogeny of the sox10 ENS phenotype. As previously shown for melanophore development, ENS progenitor fate specification fails in these mutant fish. However, in addition, we trace back the sox10 mutant ENS defect to an even earlier time point, finding that most neural crest cells fail to migrate ventrally to the gut primordium.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Sistema Nervoso Entérico/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/fisiologia , Proteínas de Homeodomínio/fisiologia , Fatores de Transcrição/fisiologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/fisiologia , Sequência de Aminoácidos , Animais , Proteínas de Transporte/metabolismo , Sistema Nervoso Central/embriologia , Mapeamento Cromossômico , Proteínas de Grupo de Alta Mobilidade/metabolismo , Proteínas de Homeodomínio/metabolismo , Hibridização In Situ , Melanóforos/metabolismo , Microscopia de Fluorescência , Modelos Genéticos , Dados de Sequência Molecular , Mutação , Crista Neural/metabolismo , Neurônios/metabolismo , Fenótipo , Filogenia , Fatores de Transcrição SOXE , Homologia de Sequência de Aminoácidos , Células-Tronco/metabolismo , Fatores de Tempo , Fatores de Transcrição/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
18.
Development ; 131(1): 241-9, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14660438

RESUMO

Components of the zebrafish GDNF receptor complex are expressed very early in the development of enteric nervous system precursors, and are already present as these cells begin to enter the gut and migrate caudally along its length. Both gfra1a and gfra1b as well as ret are expressed at this time, while gfra2 expression, the receptor component that binds the GDNF-related ligand neurturin, is not detected until the precursors have migrated along the gut. Gfra genes are also expressed in regions of the zebrafish brain and peripheral ganglia, expression domains conserved with other species. Enteric neurons are eliminated after injection with antisense morpholino oligonucleotides against ret or against both Gfra1 orthologs, but are not affected by antisense oligonucleotides against gfra2. Blocking GDNF signaling prevents migration of enteric neuron precursors, which remain positioned at the anterior end of the gut. Phenotypes induced by injection of antisense morpholinos against both Gfra orthologs can be rescued by introduction of mRNA for gfra1a or for gfra2, suggesting that GFRalpha1 and GFRalpha2 are functionally equivalent.


Assuntos
Sistema Nervoso/embriologia , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Sequência de Aminoácidos , Animais , Galinhas , Sequência Conservada , Embrião não Mamífero/fisiologia , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Humanos , Hibridização In Situ , Dados de Sequência Molecular , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas c-ret , Receptores Proteína Tirosina Quinases/química , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Proteínas de Peixe-Zebra
19.
Development ; 130(12): 2809-18, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12736222

RESUMO

The transcription factor Sox10 is required for the specification, migration and survival of all nonectomesenchymal neural crest derivatives including melanophores. sox10(-/-) zebrafish lack expression of the transcription factor mitfa, which itself is required for melanophore development. We demonstrate that the zebrafish mitfa promoter has sox10 binding sites necessary for activity in vitro, consistent with studies using mammalian cell cultures that have shown that Sox10 directly regulates Mitf expression. In addition, we demonstrate that these sites are necessary for promoter activity in vivo. We show that reintroduction of mitfa expression in neural crest cells can rescue melanophore development in sox10(-/-) embryos. This rescue of melanophores in sox10(-/-) embryos is quantitatively indistinguishable from rescue in mitfa(-/-) embryos. These findings show that the essential function of sox10 in melanophore development is limited to transcriptional regulation of mitfa. We propose that the dominant melanophore phenotype in Waardenburg syndrome IV individuals with SOX10 mutations is likely to result from failure to activate MITF in the normal number of melanoblasts.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Grupo de Alta Mobilidade/metabolismo , Melanóforos/metabolismo , Fatores de Transcrição/metabolismo , Peixe-Zebra/embriologia , Animais , Cruzamentos Genéticos , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fator de Transcrição Associado à Microftalmia , Mutação , Regiões Promotoras Genéticas , Fatores de Transcrição SOXE , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra
20.
J Mol Biol ; 320(1): 39-53, 2002 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-12079333

RESUMO

We present the results of in vitro DNA-binding assays for a mutant protein (Q44K) of the E. coli methionine repressor, MetJ, as well as the crystal structure at 2.2 A resolution of the apo-mutant bound to a 10-mer oligonucleotide encompassing an 8 bp met-box sequence. The wild-type protein binds natural operators co-operatively with respect to protein concentration forming at least a dimer of repressor dimers along operator DNAs. The minimum operator length is thus 16 bp, each MetJ dimer interacting with a single met-box site. In contrast, the Q44K mutant protein can also bind stably as a single dimer to 8 bp target sites, in part due to additional contacts made to the phosphodiester backbone outside the 8 bp target via the K44 side-chains. Protein-protein co-operativity in the mutant is reduced relative to the wild-type allowing the properties of an intermediate on the pathway to operator site saturation to be examined for the first time. The crystal structure of the decamer complex shows a unique conformation for the protein bound to the single met-box site, possibly explaining the reduced protein-protein co-operativity. In both the extended and minimal DNA complexes formed, the mutant protein makes slightly different contacts to the edges of DNA base-pairs than the wild-type, even though the site of amino acid substitution is distal from the DNA-binding motif. Quantitative binding assays suggest that this is not due to artefacts caused by the crystallisation conditions but is most likely due to the relatively small contribution of such direct contacts to the overall binding energy of DNA-protein complex formation, which is dominated by sequence-dependent distortions of the DNA duplex and by the protein-protein contact between dimers.


Assuntos
Proteínas de Bactérias/química , Escherichia coli/metabolismo , Regiões Operadoras Genéticas , Proteínas Repressoras/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Pareamento de Bases , Cristalografia por Raios X , DNA Bacteriano/genética , DNA Bacteriano/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Modelos Moleculares , Mutagênese Sítio-Dirigida , Oligonucleotídeos/química , Oligonucleotídeos/metabolismo , Ligação Proteica , Estrutura Quaternária de Proteína , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA