Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 12(9)2023 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-37174727

RESUMO

Recent studies demonstrate the adverse effects of cannabinoids on development, including via pathways shared with ethanol exposure. Our laboratory has shown that both the nervous system and cardiac development are dependent on agrin modulation of sonic hedgehog (shh) and fibroblast growth factor (Fgf) signaling pathways. As both ethanol and cannabinoids impact these signaling molecules, we examined their role on zebrafish heart development. Zebrafish embryos were exposed to a range of ethanol and/or cannabinoid receptor 1 and 2 agonist concentrations in the absence or presence of morpholino oligonucleotides that disrupt agrin or shh expression. In situ hybridization was employed to analyze cardiac marker gene expression. Exposure to cannabinoid receptor agonists disrupted midbrain-hindbrain boundary development, but had no effect on heart development, as assessed by the presence of cardiac edema or the altered expression of cardiac marker genes. In contrast, exposure to 1.5% ethanol induced cardiac edema and the altered expression of cardiac marker genes. Combined exposure to agrin or shh morpholino and 0.5% ethanol disrupted the cmlc2 gene expression pattern, with the restoration of the normal expression following shh mRNA overexpression. These studies provide evidence that signaling pathways critical to heart development are sensitive to ethanol exposure, but not cannabinoids, during early zebrafish embryogenesis.


Assuntos
Canabinoides , Peixe-Zebra , Animais , Peixe-Zebra/genética , Etanol/toxicidade , Etanol/metabolismo , Proteínas Hedgehog/metabolismo , Agrina/metabolismo , Canabinoides/metabolismo , Edema Cardíaco , Morfolinos/farmacologia , Coração
3.
J Neurosci Res ; 100(8): 1585-1601, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35014067

RESUMO

Ethanol exposure during the early stages of embryonic development can lead to a range of morphological and behavioral differences termed fetal alcohol spectrum disorders (FASDs). In a zebrafish model, we have shown that acute ethanol exposure at 8-10 hr postfertilization (hpf), a critical time of development, produces birth defects similar to those clinically characterized in FASD. Dysregulation of the Sonic hedgehog (Shh) pathway has been implicated as a molecular basis for many of the birth defects caused by prenatal alcohol exposure. We observed in zebrafish embryos that shh expression was significantly decreased by ethanol exposure at 8-10 hpf, while smo expression was much less affected. Treatment of zebrafish embryos with SAG or purmorphamine, small molecule Smoothened agonists that activate Shh signaling, ameliorated the severity of ethanol-induced developmental malformations including altered eye size and midline brain development. Furthermore, this rescue effect of Smo activation was dose dependent and occurred primarily when treatment was given after ethanol exposure. Markers of Shh signaling (gli1/2) and eye development (pax6a) were restored in embryos treated with SAG post-ethanol exposure. Since embryonic ethanol exposure has been shown to produce later-life neurobehavioral impairments, juvenile zebrafish were examined in the novel tank diving test. Our results further demonstrated that in zebrafish embryos exposed to ethanol, SAG treatment was able to mitigate long-term neurodevelopmental impairments related to anxiety and risk-taking behavior. Our results indicate that pharmacological activation of the Shh pathway at specific developmental timing markedly diminishes the severity of alcohol-induced birth defects.


Assuntos
Transtornos do Espectro Alcoólico Fetal , Efeitos Tardios da Exposição Pré-Natal , Animais , Embrião não Mamífero/metabolismo , Etanol/toxicidade , Feminino , Transtornos do Espectro Alcoólico Fetal/tratamento farmacológico , Transtornos do Espectro Alcoólico Fetal/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Gravidez , Peixe-Zebra/metabolismo
4.
Alcohol Clin Exp Res ; 44(7): 1366-1377, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32472575

RESUMO

BACKGROUND: Ethanol (EtOH) has diverse effects on nervous system development, which includes development and survival of GABAergic neurons in a sonic hedgehog (Shh) and fibroblast growth factor (Fgf)-dependent mechanism. Cannabinoids also function as inhibitors of Shh signaling, raising the possibility that EtOH and cannabinoids may interact to broadly disrupt neuronal function during brain development. METHODS: Zebrafish embryos were exposed to a range of EtOH and/or cannabinoid receptor 1 (CB1R) agonist concentrations at specific developmental stages, in the absence or presence of morpholino oligonucleotides that disrupt shh expression. In situ hybridization was employed to analyze glutamic acid decarboxylase (gad1) gene expression as a marker of GABAergic neuron differentiation, and zebrafish behavior was analyzed using the novel tank diving test as a measure of risk-taking behavior. RESULTS: Combined acute subthreshold EtOH and CB1R agonist exposure results in a marked reduction in gad1 mRNA expression in zebrafish forebrain. Consistent with the EtOH and cannabinoid effects on Shh signaling, fgf8 mRNA overexpression rescues the EtOH- and cannabinoid-induced decrease in gad1 gene expression and also prevents the changes in behavior induced by EtOH and cannabinoids. CONCLUSIONS: These studies provide evidence that forebrain GABAergic neuron development and zebrafish risk-taking behavior are sensitive to both EtOH and cannabinoid exposure in a Shh- and Fgf-dependent mechanism, and provide additional evidence that a signaling pathway involving Shh and Fgf crosstalk is a critical target of EtOH and cannabinoids in FASD.


Assuntos
Agonistas de Receptores de Canabinoides/farmacologia , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Fatores de Crescimento de Fibroblastos/genética , Neurônios GABAérgicos/efeitos dos fármacos , Proteínas Hedgehog/genética , Neurogênese/efeitos dos fármacos , Proteínas de Peixe-Zebra/genética , Animais , Comportamento Animal/efeitos dos fármacos , Embrião não Mamífero , Expressão Gênica , Glutamato Descarboxilase/efeitos dos fármacos , Glutamato Descarboxilase/genética , Proteínas Hedgehog/efeitos dos fármacos , Hibridização In Situ , Morfolinos , Neurogênese/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptor CB1 de Canabinoide/agonistas , Assunção de Riscos , Peixe-Zebra , Proteínas de Peixe-Zebra/efeitos dos fármacos
5.
Sci Rep ; 9(1): 16057, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31690747

RESUMO

We tested whether cannabinoids (CBs) potentiate alcohol-induced birth defects in mice and zebrafish, and explored the underlying pathogenic mechanisms on Sonic Hedgehog (Shh) signaling. The CBs, Δ9-THC, cannabidiol, HU-210, and CP 55,940 caused alcohol-like effects on craniofacial and brain development, phenocopying Shh mutations. Combined exposure to even low doses of alcohol with THC, HU-210, or CP 55,940 caused a greater incidence of birth defects, particularly of the eyes, than did either treatment alone. Consistent with the hypothesis that these defects are caused by deficient Shh, we found that CBs reduced Shh signaling by inhibiting Smoothened (Smo), while Shh mRNA or a CB1 receptor antagonist attenuated CB-induced birth defects. Proximity ligation experiments identified novel CB1-Smo heteromers, suggesting allosteric CB1-Smo interactions. In addition to raising concerns about the safety of cannabinoid and alcohol exposure during early embryonic development, this study establishes a novel link between two distinct signaling pathways and has widespread implications for development, as well as diseases such as addiction and cancer.


Assuntos
Canabinoides/toxicidade , Transtornos do Espectro Alcoólico Fetal/metabolismo , Proteínas Hedgehog/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Transdução de Sinais/efeitos dos fármacos , Teratogênese/efeitos dos fármacos , Animais , Etanol/efeitos adversos , Etanol/farmacologia , Feminino , Transtornos do Espectro Alcoólico Fetal/patologia , Camundongos , Receptor Smoothened/metabolismo
6.
Birth Defects Res ; 111(12): 775-788, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-30648819

RESUMO

BACKGROUND: Recent work suggests that endocannabinoids (eCBs) may signal through the sonic hedgehog signaling pathway. We therefore hypothesized that combined ethanol and eCB exposure during defined stages of zebrafish embryogenesis will produce deficits comparable to human fetal alcohol spectrum disorder (FASD). METHODS: Zebrafish embryos were exposed to ethanol or cannabinoid agonists alone or in combination at defined developmental stages and assessed for changes in brain morphology or expression of marker genes such as pax6a. Juvenile fish were then assessed for risk-taking/anxiety-like behavior using the novel tank dive test. RESULTS: Either chronic or acute exposure to high doses of the CB1R agonist ACEA resulted in FASD phenotypes. However, acute subthreshold doses of CB1R agonist alone, or combined with 0.5% ethanol, did not induce morphological phenotypes, but did induce dysmorphogenesis when combined with acute 1% ethanol. Phenotypes were rescued using the CB1R antagonist SR141716A. In addition, JZL195, a dual inhibitor of FAAH and MAGL, two degradative enzymes for eCBs, induced FASD phenotypes in the presence of subthreshold ethanol, confirming the activation of common signaling pathways by ethanol and eCBs. We next analyzed the effects of ethanol and CB1R agonist on juvenile zebrafish behavior and show that ACEA or ethanol alone did not alter behavior, but combined ACEA and ethanol increased risk-taking behavior. CONCLUSIONS: These studies demonstrate that pathological and behavioral phenotypes associated with FASD are induced by exposure to CB1R agonists and suggest that combined exposure to lower levels of alcohol and marijuana may be capable of inducing FASD-like morphological and behavioral impairments.


Assuntos
Canabinoides/efeitos adversos , Embrião não Mamífero/embriologia , Etanol/efeitos adversos , Transtornos do Espectro Alcoólico Fetal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteínas de Peixe-Zebra/biossíntese , Peixe-Zebra/embriologia , Animais , Canabinoides/farmacologia , Modelos Animais de Doenças , Embrião não Mamífero/patologia , Etanol/farmacologia , Transtornos do Espectro Alcoólico Fetal/patologia , Transtornos do Espectro Alcoólico Fetal/fisiopatologia
7.
Exp Brain Res ; 235(8): 2413-2423, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28493069

RESUMO

This study was undertaken to ascertain whether defined markers of early zebrafish brain development are affected by chronic ethanol exposure or morpholino knockdown of agrin, sonic hedgehog, retinoic acid, and fibroblast growth factors, four signaling molecules that are suggested to be ethanol sensitive. Zebrafish embryos were exposed to 2% ethanol from 6 to 24 hpf or injected with agrin, shha, aldh1a3, or fgf8a morpholinos. In situ hybridization was employed to analyze otx2, pax6a, epha4a, krx20, pax2a, fgf8a, wnt1, and eng2b expression during early brain development. Our results showed that pax6a mRNA expression was decreased in eye, forebrain, and hindbrain of both chronic ethanol exposed and select MO treatments. Epha4a expression in rhombomere R1 boundary was decreased in chronic ethanol exposure and aldh1a3 morphants, lost in fgf8a morphants, but largely unaffected in agrin and shha morphants. Ectopic pax6a and epha4a expression in midbrain was only found in fgf8a morphants. These results suggest that while chronic ethanol induces obvious morphological change in brain architecture, many molecular markers of these brain structures are relatively unaffected by ethanol exposure.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteínas de Peixe-Zebra/metabolismo , Fatores Etários , Animais , Encéfalo/anatomia & histologia , Encéfalo/embriologia , Embrião de Mamíferos , Olho/efeitos dos fármacos , Olho/embriologia , Olho/metabolismo , Hibridização In Situ , Morfolinas/farmacologia , Tretinoína/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
8.
Neurotoxicol Teratol ; 61: 66-73, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28223149

RESUMO

BACKGROUND: Developmental exposure to ethanol is recognized to produce long-term neurobehavioral impairment in multiple animal models. However, the molecular mechanisms underlying these deficits remain poorly understood. The present study was undertaken to ascertain whether two well-characterized targets of prenatal alcohol exposure, sonic hedgehog (Shh) and retinoic acid (RA), that induce the hallmark morphological phenotypes of fetal alcohol spectrum disorders (FASD), are involved in the generation of behavioral alterations as a result of alcohol exposure. METHODS: Zebrafish embryos were exposed to ethanol (0%, 1%, 3%) at either 8-10 or 24-27h post-fertilization (hpf) and then evaluated during adolescence in the novel tank dive test to assess anxiety and risk-taking behavior. Overt signs of dysmorphogenesis were also scored and behavioral and morphological changes were compared for embryos treated with alcohol alone or in combination with subthreshold doses of shh or alhh1a3 morpholinos (MOs). RESULTS: Ethanol treated fish displayed altered tank diving behavior that was not exacerbated by combined MO treatment. While treatment of embryos with either shha mRNA or RA prior to ethanol exposure only ameliorated the altered tank diving response in the case of shha mRNA overexpression, dysmorphogenesis was rescued by both treatments. CONCLUSION: These results suggest that the effects of ethanol exposure on changes in anxiety and risk-taking behavior in adolescent zebrafish is manifested by a blunting of Shh, but not RA, signaling during early development.


Assuntos
Comportamento Animal/efeitos dos fármacos , Etanol/toxicidade , Proteínas Hedgehog/fisiologia , Morfolinos/farmacologia , Tretinoína , Proteínas de Peixe-Zebra/fisiologia , Animais , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Feminino , Masculino , Oligonucleotídeos Antissenso/farmacologia , Gravidez , Peixe-Zebra
9.
Birth Defects Res A Clin Mol Teratol ; 103(12): 1046-57, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26470995

RESUMO

BACKGROUND: Ethanol is a teratogen affecting numerous regions of the developing nervous system. The present study was undertaken to ascertain whether ethanol independently disrupts distinct signaling pathways or rather disrupts interactive pathways that regulate development of ethanol-sensitive tissues. METHODS: Zebrafish embryos were exposed to ethanol in the absence or presence of aldh1a3 or Shh morpholino oligonucleotides (MOs), which disrupt retinoic acid (RA) or sonic hedgehog (Shh) function, respectively. Morphological analysis of ocular or midbrain-hindbrain boundary (MHB) development was conducted, and the ability to rescue ethanol and MO-induced phenotypes was assessed. In situ hybridization was used to analyze Pax6a expression during ocular development. RESULTS: Chronic ethanol exposure, or combined ethanol and MO treatment, results in perturbed MHB formation and microphthalmia. While RA can rescue the MHB phenotype following ethanol combined with either MO, Shh mRNA is unable to rescue the disrupted MHB with combined ethanol and aldh1a3 MO treatment. RA also is unable to rescue microphthalmia induced by ethanol and Shh MO. CONCLUSION: These studies demonstrate that while reduction of either RA or Shh signaling produces the same disruption of MHB or ocular development, that can be phenocopied using ethanol combined with either MO, RA overexpression can only rescue disrupted MHB, but not microphthalmia, in combined subthreshold Shh MO and ethanol. Our data suggest that MHB development may involve crosstalk between RA and Shh signaling, while ocular development depends on RA and Shh signaling that both are targets of ethanol in fetal alcohol spectrum disorders but do not depend on a mechanism involving crosstalk.


Assuntos
Etanol/toxicidade , Proteínas Hedgehog/metabolismo , Tretinoína/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Encéfalo/embriologia , Olho/embriologia , Proteínas do Olho/genética , Proteínas de Homeodomínio/genética , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Proteínas Repressoras/genética , Transdução de Sinais/efeitos dos fármacos
10.
Neurotoxicol Teratol ; 44: 70-80, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24929233

RESUMO

Alcohol is a teratogen that has diverse effects on brain and craniofacial development, leading to a constellation of developmental disorders referred to as fetal alcohol spectrum disorder (FASD). The molecular basis of ethanol insult remains poorly understood, as does the relationship between molecular and behavioral changes as a consequence of prenatal ethanol exposure. Zebrafish embryos were exposed to a range of ethanol concentrations (0.5-5.0%) during defined developmental stages, and examined for morphological phenotypes characteristic of FASD. Embryos were also analyzed by in situ hybridization for changes in expression of defined cell markers for neural cell types that are sonic hedgehog-dependent. We show that transient binge-like ethanol exposures during defined developmental stages, such as early gastrulation and early neurulation, result in a range of phenotypes and changes in expression of Shh-dependent genes. The severity of fetal alcohol syndrome (FAS) morphological phenotypes, such as microphthalmia, depends on the embryonic stage and concentration of alcohol exposure, as does diminution of retinal Pax6a or forebrain and hindbrain GAD1 gene expression. We also show that changes in eye and brain morphology correlate with changes in Pax6a and GAD1 gene expression. Our results therefore show that transient binge-like ethanol exposures in zebrafish embryos produce the stereotypical morphological phenotypes of FAS, with the severity of phenotypes depending on the developmental stage and alcohol concentration of exposure.


Assuntos
Embrião não Mamífero/efeitos dos fármacos , Etanol/toxicidade , Transtornos do Espectro Alcoólico Fetal/genética , Transtornos do Espectro Alcoólico Fetal/patologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Animais , Proteínas do Olho/genética , Feminino , Glutamato Descarboxilase/genética , Proteínas de Homeodomínio/genética , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Gravidez , Proteínas Repressoras/genética , Peixe-Zebra/embriologia
11.
Zebrafish ; 10(2): 218-27, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23672288

RESUMO

Cdx2 has been suggested to play an important role in Barrett's esophagus or intestinal metaplasia (IM) in the esophagus. To investigate whether transgenic overexpression of cdx1b, the functional equivalent of mammalian Cdx2 in zebrafish, may lead to IM of zebrafish esophageal squamous epithelium, a transgenic zebrafish system was developed by expressing cdx1b gene under the control of zebrafish keratin 5 promoter (krt5p). Gene expression in the esophageal squamous epithelium of wild-type and transgenic zebrafish was analyzed by Affymetrix microarray and confirmed by in situ hybridization. Morphology, mucin expression, cell proliferation, and apoptosis were analyzed by hematoxylin & eosin (HE) staining, Periodic acid Schiff (PAS) Alcian blue staining, proliferating cell nuclear antigen (PCNA) immunohistochemical staining, and TUNEL assay as well. cdx1b was found to be overexpressed in the nuclei of esophageal squamous epithelial cells of the transgenic zebrafish. Ectopic expression of cdx1b disturbed the development of this epithelium in larval zebrafish and induced metaplastic changes in gene expression in the esophageal squamous epithelial cells of adult zebrafish, that is, up-regulation of intestinal differentiation markers and down-regulation of squamous differentiation markers. However, cdx1b failed to induce histological IM, or to modulate cell proliferation and apoptosis in the squamous epithelium of adult transgenic zebrafish.


Assuntos
Esôfago de Barrett/genética , Esôfago/patologia , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Modelos Animais , Proteínas de Peixe-Zebra/genética , Peixe-Zebra , Animais , Esôfago de Barrett/metabolismo , Esôfago de Barrett/fisiopatologia , Proliferação de Células , Epitélio/metabolismo , Epitélio/patologia , Esôfago/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Hibridização In Situ , Queratina-5/genética , Queratina-5/metabolismo , Larva , Metaplasia/genética , Metaplasia/metabolismo , Metaplasia/patologia , Mucinas/genética , Mucinas/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Proteínas de Peixe-Zebra/metabolismo
12.
Artigo em Inglês | MEDLINE | ID: mdl-23184466

RESUMO

BACKGROUND: Ethanol is a teratogen that affects numerous developmental processes in the nervous system, which includes development and survival of GABAergic and glutamatergic neurons. Possible molecular mechanisms accounting for ethanol's effects on nervous system development include perturbed fibroblast growth factor (Fgf) and Sonic hedgehog (Shh) signaling. In zebrafish, forebrain GABAergic neuron development is dependent on Fgf19 and Shh signaling. The present study was conducted to test the hypothesis that ethanol affects GABAergic and glutamatergic neuron development by disrupting Fgf, Shh, and agrin function. METHODS: Zebrafish embryos were exposed to varying concentrations of ethanol during a range of developmental stages, in the absence or presence of morpholino oligonucleotides (MOs) that disrupt agrin or Shh function. In situ hybridization was used to analyze glutamic acid decarboxylase (GAD1) gene expression, as well as markers of glutamatergic neurons. RESULTS: Acute ethanol exposure results in marked reduction in GAD1 gene expression in forebrain and hindbrain, and reduction of glutamatergic neuronal markers in hindbrain. Subthreshold ethanol exposure, combined with agrin or Shh MO treatment, produces a similar diminution in expression of markers for GABAergic and glutamatergic neurons. Consistent with the ethanol effects on Fgf and Shh pathways, Fgf19, Fgf8, or Shh mRNA overexpression rescues ethanol-induced decreases in GAD1 and Atonal1a gene expression. CONCLUSIONS: These studies demonstrate that GABAergic and glutamatergic neuron development in zebrafish forebrain or cerebellum is sensitive to ethanol exposure, and provides additional evidence that a signaling pathway involving agrin, Fgfs and Shh may be a critical target of ethanol exposure during zebrafish embryogenesis.


Assuntos
Encéfalo/efeitos dos fármacos , Depressores do Sistema Nervoso Central/toxicidade , Desenvolvimento Embrionário/efeitos dos fármacos , Etanol/toxicidade , Teratogênicos/toxicidade , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Agrina/genética , Agrina/metabolismo , Animais , Biomarcadores/metabolismo , Encéfalo/embriologia , Encéfalo/metabolismo , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Inativação Gênica , Glutamato Descarboxilase/genética , Ácido Glutâmico/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Morfolinos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/embriologia , Prosencéfalo/metabolismo , Rombencéfalo/efeitos dos fármacos , Rombencéfalo/embriologia , Rombencéfalo/metabolismo , Transdução de Sinais
13.
Int Rev Cell Mol Biol ; 299: 255-315, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22959306

RESUMO

Alcohol (ethanol) is a teratogen that adversely affects nervous system development in a wide range of animal species. In humans numerous congenital abnormalities arise as a result of fetal alcohol exposure, leading to a spectrum of disorders referred to as fetal alcohol spectrum disorder (FASD). These abnormalities include craniofacial defects as well as neurological defects that affect a variety of behaviors. These human FASD phenotypes are reproduced in the rodent central nervous system (CNS) following prenatal ethanol exposure. While the study of ethanol effects on zebrafish development has been more limited, several studies have shown that different strains of zebrafish exhibit differential susceptibility to ethanol-induced cyclopia, as well as behavioral deficits. Molecular mechanisms underlying the effects of ethanol on CNS development also appear to be shared between rodent and zebrafish. Thus, zebrafish appear to recapitulate the observed effects of ethanol on human and mouse CNS development, indicating that zebrafish can serve as a complimentary developmental model system to study the molecular basis of FASD. Recent studies examining the effect of ethanol exposure on zebrafish nervous system development are reviewed, with an emphasis on attempts to elucidate possible molecular pathways that may be impacted by developmental ethanol exposure. Recent work from our laboratories supports a role for perturbed extracellular matrix function in the pathology of ethanol exposure during zebrafish CNS development. The use of the zebrafish model to assess the effects of ethanol exposure on adult nervous system function as manifested by changes in zebrafish behavior is also discussed.


Assuntos
Etanol/toxicidade , Sistema Nervoso/efeitos dos fármacos , Sistema Nervoso/embriologia , Peixe-Zebra/embriologia , Animais , Modelos Animais de Doenças , Feminino , Transtornos do Espectro Alcoólico Fetal/fisiopatologia , Neurônios Motores/citologia , Neurônios Motores/efeitos dos fármacos , Sistema Nervoso/citologia , Vias Neurais/citologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/embriologia , Neurogênese/efeitos dos fármacos , Gravidez
14.
Cell ; 146(1): 164-76, 2011 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-21729787

RESUMO

The mechanisms that generate specific neuronal connections in the brain are under intense investigation. In zebrafish, retinal ganglion cells project their axons into at least six layers within the neuropil of the midbrain tectum. Each axon elaborates a single, planar arbor in one of the target layers and forms synapses onto the dendrites of tectal neurons. We show that the laminar specificity of retinotectal connections does not depend on self-sorting interactions among RGC axons. Rather, tectum-derived Slit1, signaling through axonal Robo2, guides neurites to their target layer. Genetic and biochemical studies indicate that Slit binds to Dragnet (Col4a5), a type IV Collagen, which forms the basement membrane on the surface of the tectum. We further show that radial glial endfeet are required for the basement-membrane anchoring of Slit. We propose that Slit1 signaling, perhaps in the form of a superficial-to-deep gradient, presents laminar positional cues to ingrowing retinal axons.


Assuntos
Encéfalo/embriologia , Colágeno Tipo IV/metabolismo , Teto do Mesencéfalo/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Axônios/metabolismo , Encéfalo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores Imunológicos/metabolismo , Células Ganglionares da Retina/metabolismo , Transdução de Sinais , Peixe-Zebra/metabolismo
15.
Birth Defects Res A Clin Mol Teratol ; 91(3): 129-41, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21308976

RESUMO

BACKGROUND: Alcohol (ethanol) is a teratogen known to affect the developing eyes, face, and brain. Among the ocular defects in fetal alcohol spectrum disorder (FASD) are microphthalmia and optic nerve hypoplasia. Employing zebrafish as an FASD model provides an excellent system to analyze the molecular basis of prenatal ethanol exposure-induced defects because embryos can be exposed to ethanol at defined developmental stages and affected genetic pathways can be examined. We have previously shown that disruption of agrin function in zebrafish embryos produces microphthalmia and optic nerve hypoplasia. METHODS: Zebrafish embryos were exposed to varying concentrations of ethanol in the absence or presence of morpholino oligonucleotides (MOs) that disrupt agrin function. In situ hybridization was used to analyze ocular gene expression as a consequence of ethanol exposure and agrin knockdown. Morphologic analysis of zebrafish embryos was also conducted. RESULTS: Acute ethanol exposure induces diminished agrin gene expression in zebrafish eyes and, importantly, combined treatment with subthreshold levels of agrin MO and ethanol produces pronounced microphthalmia, markedly reduces agrin gene expression, and perturbs Pax6a and Mbx gene expression. Microphthalmia produced by combined agrin MO and ethanol treatment was rescued by sonic hedgehog (Shh) mRNA overexpression, suggesting that ethanol-mediated disruption of agrin expression results in disrupted Shh function. CONCLUSIONS: These studies illustrate the strong potential for using zebrafish as a model to aid in defining the molecular basis for ethanol's teratogenic effects. The results of this work suggest that agrin expression and function may be a target of ethanol exposure during embryogenesis.


Assuntos
Agrina/fisiologia , Etanol/farmacologia , Olho/efeitos dos fármacos , Olho/embriologia , Peixe-Zebra/embriologia , Agrina/genética , Agrina/metabolismo , Animais , Animais Geneticamente Modificados , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Embrião não Mamífero , Exposição Ambiental , Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/fisiologia , Microftalmia/induzido quimicamente , Microftalmia/genética , Microftalmia/patologia , Oligorribonucleotídeos Antissenso/farmacologia , Fenótipo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
16.
Dev Dyn ; 239(11): 3038-47, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20931647

RESUMO

Transitin is a nestin-like intermediate filament protein co-expressed with vimentin in the precursor cells of the myogenic and neurogenic lineages of the avian embryo. To understand its role in myogenesis, stable cell lines expressing transitin-targeted siRNAs were derived from the quail muscle cell line QM7. When cells were cultured in differentiation medium, we found that transitin knockdown prevented myoblast fusion and myotube formation. MyoD mRNA could be detected in transitin siRNA-transfected cells, but upregulation of myogenin and desmin expression was impaired compared to control cells. In addition, transitin siRNA cells maintain high levels of Pax7 expression suggesting that QM7 myoblasts into which transitin expression has been attenuated display a muscle progenitor cell phenotype (Pax7(+)/MyoD(+)/myogenin(-)/desmin(-)). These observations indicate that transitin plays an important role in the initiation of the myogenic program in avian muscle progenitor cells in acting downstream of MyoD and upstream of myogenin during the lineage progression.


Assuntos
Proteínas Aviárias/metabolismo , Diferenciação Celular/fisiologia , Proteínas de Filamentos Intermediários/metabolismo , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Animais , Proteínas Aviárias/genética , Diferenciação Celular/genética , Linhagem Celular , Imunofluorescência , Immunoblotting , Proteínas de Filamentos Intermediários/genética , Codorniz , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Matrix Biol ; 29(5): 402-10, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20362054

RESUMO

Basement membranes (BMs) are considered to be uniform, approximately 100 nm-thin extracellular matrix sheets that serve as a substrate for epithelial cells, endothelial cells and myotubes. To find out whether BMs maintain their ultrastructure, protein composition and biophysical properties throughout life the natural aging history of the human inner limiting membranes (ILM) was investigated. The ILM is a BM at the vitreal surface of the retina that connects the retina with the vitreous. Transmission electron microscopy (TEM) showed that the ILM steadily increases in thickness from 70 nm at fetal stages to several microns at age 90. By the age of 20, the ILM loses its laminated structure to become an amorphous and very irregular extracellular matrix layer. Atomic force microscopy (AFM) showed that the native, hydrated ILMs are on average 4-fold thicker than the dehydrated ILMs as seen by TEM and that their thickness is prominently determined by its water-binding proteoglycans. The morphological changes are accompanied by age-related changes in the biochemical composition, whereby the relative concentrations of collagen IV and agrin increase, and the concentration of laminin decreases with age. Force-indentation measurements by AFM also showed that ILMs become increasingly stiffer with advancing age. The data suggest that BMs from other human tissues may undergo similar age-related changes.


Assuntos
Membrana Basal/fisiologia , Colágeno Tipo IV/fisiologia , Laminina/fisiologia , Retina/fisiologia , Adulto , Fatores Etários , Membrana Basal/química , Membrana Basal/ultraestrutura , Western Blotting , Feminino , Feto , Humanos , Imuno-Histoquímica , Masculino , Microscopia de Força Atômica , Microscopia Eletrônica de Transmissão , Retina/química , Retina/ultraestrutura
18.
Oral Oncol ; 46(1): 31-7, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19932050

RESUMO

The zebrafish has become a useful model organism for research on development and diseases. However, there has been no zebrafish model system for studying oral carcinogenesis. In the present study, we first characterized the histology of the upper gastrointestinal tract of zebrafish. We found that zebrafish tongue was covered by a non-keratinized stratified squamous epithelium, which was similar to the oro-esophageal epithelium in humans. In situ hybridization showed that keratin 5, a marker of the basal cell layer of mammalian oral epithelium, was expressed in the squamous epithelium of zebrafish tongue. A highly conserved promoter of zebrafish keratin 5 was cloned to drive transgenic expression of GFP. GFP was found to be expressed in the periderm of embryos. In adult fish, GFP was also abundantly expressed in the tongue and fin. GFP expression in transgenic fish recapitulated endogenous zebrafish keratin 5 gene expression as shown by in situ hybridization. This study indicated a high fidelity of GFP reporter gene expression in the tongue under the control of zebrafish keratin 5 promoter. This zebrafish transgenic model system may be used for future studies on oral development and cancer.


Assuntos
Trato Gastrointestinal/anatomia & histologia , Proteínas de Fluorescência Verde/genética , Queratina-5/genética , Regiões Promotoras Genéticas/genética , Língua/anatomia & histologia , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Pesquisa Biomédica , Regulação Neoplásica da Expressão Gênica/genética , Proteínas de Fluorescência Verde/metabolismo , Queratina-15/genética , Queratina-15/metabolismo , Queratina-5/metabolismo , Modelos Animais , Dados de Sequência Molecular , Peixe-Zebra/anatomia & histologia
19.
Dev Neurobiol ; 68(7): 877-98, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18327763

RESUMO

Recent studies from our laboratory have begun to elucidate the role of agrin in zebrafish development. One agrin morphant phenotype that results from agrin knockdown is microphthalmia (reduced eye size). To begin to understand the mechanisms underlying the role of agrin in eye development, we have analyzed retina development in agrin morphants. Retinal differentiation is impaired in agrin morphants, with retinal lamination being disrupted following agrin morpholino treatment. Pax 6.1 and Mbx1 gene expression, markers of eye development, are markedly reduced in agrin morphants. Formation of the optic fiber layer of the zebrafish retina is also impaired, exhibited as both reduced size of the optic fiber layer, and disruption of retinal ganglion cell axon growth to the optic tectum. The retinotectal topographic projection to the optic tectum is perturbed in agrin morphants in association with a marked loss of heparan sulfate expression in the retinotectal pathway, with this phenotype resembling retinotectal phenotypes observed in mutant zebrafish lacking enzymes for heparan sulfate synthesis. Treatment of agrin morphants with a fibroblast growth factor (Fgf) receptor inhibitor, rescue of the retinal lamination phenotype by transplantation of Fgf8-coated beads, and disruption of both the expression of Fgf-dependent genes and activation of ERK in agrin morphants provides evidence that agrin modulation of Fgf function contributes to retina development. Collectively, these agrin morphant phenotypes provide support for a crucial role of agrin in retina development and formation of an ordered retinotectal topographic map in the optic tectum of zebrafish.


Assuntos
Agrina/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteoglicanas de Heparan Sulfato/fisiologia , Retina/embriologia , Retina/crescimento & desenvolvimento , Peixe-Zebra , Laranja de Acridina , Animais , Animais Geneticamente Modificados , Morte Celular , Proteínas ELAV/genética , Proteína Semelhante a ELAV 3 , Embrião não Mamífero/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Fator 3 de Crescimento de Fibroblastos/genética , Fator 3 de Crescimento de Fibroblastos/metabolismo , Proteína GAP-43/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Oligonucleotídeos Antissenso/farmacologia , Pirróis/farmacologia , Retina/efeitos dos fármacos , Retina/metabolismo , Esfingosina/análogos & derivados , Colículos Superiores/embriologia , Colículos Superiores/crescimento & desenvolvimento , Colículos Superiores/metabolismo , Fator de Transcrição Brn-3C/genética , Peixe-Zebra/embriologia , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
20.
Mech Dev ; 125(1-2): 167-81, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18037275

RESUMO

Olfactomedins comprise a diverse family of secreted glycoproteins, which includes noelin, tiarin, pancortin and gliomedin, implicated in development of the nervous system, and the glaucoma-associated protein myocilin. Here we show in zebrafish that olfactomedin-2 (OM2) is a developmentally regulated gene, and that knockdown of protein expression by morpholino antisense oligonucleotides leads to perturbations of nervous system development. Interference with OM2 expression results in impaired development of branchiomotor neurons, specific disruption of the late phase branchiomotor axon guidance, and affects development of the caudal pharyngeal arches, olfactory pits, eyes and optic tectum. Effects of OM2 knockdown on eye development are likely associated with Pax6 signaling in developing eyes, as Pax6.1 and Pax6.2 mRNA expression patterns are altered in the eyes of OM2 morphants. The specific absence of most cartilaginous structures in the pharyngeal arches indicates that the observed craniofacial phenotypes may be due to perturbed differentiation of cranial neural crest cells. Our studies show that this member of the olfactomedin protein family is an important regulator of development of the anterior nervous system.


Assuntos
Padronização Corporal , Sistema Nervoso Central/embriologia , Proteínas da Matriz Extracelular/fisiologia , Glicoproteínas/fisiologia , Peixe-Zebra/embriologia , Animais , Proteínas da Matriz Extracelular/genética , Proteínas do Olho/genética , Regulação da Expressão Gênica no Desenvolvimento , Glicoproteínas/genética , Proteínas de Homeodomínio/genética , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , RNA Mensageiro/genética , Proteínas Repressoras/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...