Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros













Base de dados
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Iran J Immunol ; 12(2): 94-103, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26119192

RESUMO

BACKGROUND: During the initial phase of an infection, there is an upregulation of inducible nitric oxide synthase in the macrophages for the production of nitric oxide. This is followed by the recruitment of polymorphonuclear leukocytes (neutrophils) which release arginase. Arginase competes with inducible nitric oxide synthase for a common substrate L-arginine. OBJECTIVE: To investigate whether the entry of neutrophils and release of arginase can interfere with nitric oxide production from stimulated mouse macrophages. METHODS: Neutrophils were isolated from human blood and stimulated with cytodex-3 beads. Cultured macrophages were stimulated with lipopolysaccharide and interferon gamma with or without N (G)-nitro-L-arginine methyl ester or N (omega)-hydroxy-nor-L-arginine. Measurement of NO2-/NO3- and urea were done using the spectrophotometer. RESULTS: A significantly higher level of nitric oxide production from stimulated macrophages was observed compared to control. There was a decrease in nitric oxide production when stimulated macrophages were treated with the supernatant from activated neutrophils (p<0.05). CONCLUSION: Arginase from neutrophils can modulate nitric oxide production from activated macrophages which may affect the course of infection by intracellular bacteria.


Assuntos
Arginase/metabolismo , Macrófagos/imunologia , Neutrófilos/imunologia , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico/biossíntese , Animais , Arginase/biossíntese , Arginina/análogos & derivados , Arginina/farmacologia , Linhagem Celular , Dextranos/imunologia , Humanos , Interferon gama , Lipopolissacarídeos , Ativação de Macrófagos/imunologia , Camundongos , Mycobacterium tuberculosis/imunologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Nitroarginina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA