Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Am Chem Soc ; 142(38): 16240-16253, 2020 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-32866014

RESUMO

This Article outlines the optimized chemical synthesis and preliminary biochemical characterization of a new oligonucleotide analogue called thiophosphoramidate morpholinos (TMOs). Their rational design hinges upon integrating two well-studied pharmacophores, namely, phosphorothioates (pS) and morpholinos, to create morpholino-pS hybrid oligonucleotides. Our simple synthesis strategy enables the easy incorporation of morpholino-pS moieties and therapeutically relevant sugar modifications in tandem to create novel oligonucleotide (ON) analogues that are hitherto unexplored in the oligotherapeutics arena. Exclusively TMO-modified ONs demonstrate high stability toward 3'-exonuclease. Hybridization studies show that TMO chimeras consisting of alternating TMO and DNA-pS subunits exhibit higher binding affinity toward complementary RNA relative to the canonical DNA/RNA duplex (∼10 °C). Oligonucleotides that consist entirely of TMO linkages also show higher RNA binding affinity but do not recruit ribonuclease H1 (RNase H1). Chimeric TMO analogues demonstrate high gene silencing efficacy, comparable to that of a chimeric 2'-OMe-pS/pO control, during in vitro bioassay screens designed to evaluate their potential as microRNA inhibitors of hsa-miR-15b-5p in HeLa cells.


Assuntos
MicroRNAs/antagonistas & inibidores , Morfolinos/farmacologia , Fosfatos/farmacologia , Células HeLa , Humanos , Estrutura Molecular , Morfolinos/síntese química , Morfolinos/química , Fosfatos/síntese química , Fosfatos/química
2.
Peptides ; 119: 170080, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31260713

RESUMO

Nesfatin-1 is an anorexic peptide derived from nucleobindin 2 (NUCB2). An increase in hypothalamic nesfatin-1 inhibits feeding behavior and promotes weight loss. However, the effects of weight loss on hypothalamic nesfatin-1 levels are unclear. In this study, obese rats lost weight in three ways: Calorie Restriction diet (CRD), Sleeve gastrectomy (SG) and Roux-en-Y gastric bypass (RYGB). We found an increase in nesfatin-1 serum and cerebrospinal fluid levels after weight loss in obese Sprague-Dawley (SD) rats. Moreover, weight loss also increased hypothalamic melanocortin 3/4 receptor (MC3/4R) and extracellular regulated kinase phosphorylation (p-ERK) signaling. Third ventricle administration of antisense morpholino oligonucleotide (MON) against the gene encoding NUCB2 inhibited hypothalamic nesfatin-1 and p-ERK signaling, increased food intake and reduced body weight loss in SG and RYGB obese rats. Third ventricle administration of SHU9119 (MC3/4R blocker) blocked hypothalamic MC3/4R, inhibited p-ERK signaling, increased food intake and reduced body weight loss in SG and RYGB obese rats. These findings indicate that weight loss leads to an increase in hypothalamic nesfatin-1. The increase in hypothalamic nesfatin-1 participates in regulating feeding behavior through the MC3/4R-ERK signaling especially after SG and RYGB.


Assuntos
Comportamento Alimentar , Hipotálamo/metabolismo , Sistema de Sinalização das MAP Quinases , Nucleobindinas/metabolismo , Obesidade/metabolismo , Receptor Tipo 3 de Melanocortina/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Animais , Hipotálamo/patologia , Masculino , Morfolinos/genética , Morfolinos/farmacologia , Nucleobindinas/antagonistas & inibidores , Nucleobindinas/genética , Obesidade/genética , Obesidade/patologia , Ratos , Ratos Sprague-Dawley , Receptor Tipo 3 de Melanocortina/genética , Receptor Tipo 4 de Melanocortina/genética
3.
J Alzheimers Dis ; 63(3): 1173-1189, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29710707

RESUMO

Oxidative stress and amyloid-ß (Aß) oligomers have been implicated in Alzheimer's disease (AD). The growth and maintenance of neuronal networks are influenced by brain derived neurotrophic factor (BDNF) expression, which is promoted by protein kinase C epsilon (PKCɛ). We investigated the reciprocal interaction among oxidative stress, Aß, and PKCɛ levels and subsequent PKCɛ-dependent MnSOD and BDNF expression in hippocampal pyramidal neurons. Reduced levels of PKCɛ, MnSOD, and BDNF and an increased level of Aß were also found in hippocampal neurons from autopsy-confirmed AD patients. In cultured human primary hippocampal neurons, spherical aggregation of Aß (amylospheroids) decreased PKCɛ and MnSOD. Treatment with t-butyl hydroperoxide (TBHP) increased superoxide, the oxidative DNA/RNA damage marker, 8-OHG, and Aß levels, but reduced PKCɛ, MnSOD, BDNF, and cultured neuron density. These changes were reversed with the PKCɛ activators, bryostatin and DCPLA-ME. PKCɛ knockdown suppressed PKCɛ, MnSOD, and BDNF but increased Aß. In cultured neurons, the increase in reactive oxygen species (ROS) associated with reduced PKCɛ during neurodegeneration was inhibited by the SOD mimetic MnTMPyP and the ROS scavenger NAc, indicating that strong oxidative stress suppresses PKCɛ level. Reduction of PKCɛ and MnSOD was prevented with the PKCɛ activator bryostatin in 5-6-month-old Tg2576 AD transgenic mice. In conclusion, oxidative stress and Aß decrease PKCɛ expression. Reciprocally, a depression of PKCɛ reduces BDNF and MnSOD, resulting in oxidative stress. These changes can be prevented with the PKCɛ-specific activators.


Assuntos
Doença de Alzheimer/patologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Regulação para Baixo/fisiologia , Hipocampo/patologia , Neurônios/metabolismo , Proteína Quinase C-épsilon/deficiência , Adjuvantes Imunológicos/farmacologia , Idoso , Idoso de 80 Anos ou mais , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Animais , Briostatinas/metabolismo , Briostatinas/farmacologia , Células Cultivadas , Feminino , Feto/anatomia & histologia , Hipocampo/citologia , Hipocampo/metabolismo , Humanos , Masculino , Metaloporfirinas/farmacologia , Camundongos , Pessoa de Meia-Idade , Morfolinos/farmacologia , Proteína Quinase C-épsilon/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Transfecção , terc-Butil Hidroperóxido/farmacologia
4.
J Med Chem ; 61(3): 1086-1097, 2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29300474

RESUMO

A novel series of morpholine-based nonsteroidal mineralocorticoid receptor antagonists is reported. Starting from a pyrrolidine HTS hit 9 that possessed modest potency but excellect selectivity versus related nuclear hormone receptors, a series of libraries led to identification of morpholine lead 10. After further optimization, cis disubstituted morpholine 22 was discovered, which showed a 45-fold boost in binding affinity and corresponding functional potency compared to 13. While 22 had high clearance in rat, it provided sufficient exposure at high doses to favorably assess in vivo efficacy (increased urinary Na+/K+ ratio) and safety. In contrast to rat, the dog and human MetID and PK profiles of 22 were adequate, suggesting that it could be suitable as a potential clinical asset.


Assuntos
Antagonistas de Receptores de Mineralocorticoides/química , Antagonistas de Receptores de Mineralocorticoides/farmacologia , Morfolinos/química , Morfolinos/farmacologia , Oxazinas/química , Receptores de Mineralocorticoides/metabolismo , Animais , Ensaios Clínicos Fase I como Assunto , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Concentração Inibidora 50 , Modelos Moleculares , Conformação Proteica , Ratos , Ratos Wistar , Receptores de Mineralocorticoides/química , Relação Estrutura-Atividade
5.
Development ; 144(8): 1518-1530, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28255006

RESUMO

Folate supplementation prevents up to 70% of neural tube defects (NTDs), which result from a failure of neural tube closure during embryogenesis. The elucidation of the mechanisms underlying folate action has been challenging. This study introduces Xenopus laevis as a model to determine the cellular and molecular mechanisms involved in folate action during neural tube formation. We show that knockdown of folate receptor 1 (Folr1; also known as FRα) impairs neural tube formation and leads to NTDs. Folr1 knockdown in neural plate cells only is necessary and sufficient to induce NTDs. Folr1-deficient neural plate cells fail to constrict, resulting in widening of the neural plate midline and defective neural tube closure. Pharmacological inhibition of folate action by methotrexate during neurulation induces NTDs by inhibiting folate interaction with its uptake systems. Our findings support a model in which the folate receptor interacts with cell adhesion molecules, thus regulating the apical cell membrane remodeling and cytoskeletal dynamics necessary for neural plate folding. Further studies in this organism could unveil novel cellular and molecular events mediated by folate and lead to new ways of preventing NTDs.


Assuntos
Polaridade Celular , Receptor 1 de Folato/metabolismo , Placa Neural/citologia , Placa Neural/metabolismo , Tubo Neural/citologia , Tubo Neural/embriologia , Organogênese , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriologia , Animais , Caderinas/metabolismo , Polaridade Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Endocitose/efeitos dos fármacos , Feminino , Receptor 1 de Folato/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Marcação de Genes , Humanos , Morfolinos/farmacologia , Tubo Neural/metabolismo , Neurulação/efeitos dos fármacos , Organogênese/efeitos dos fármacos , Proteínas de Xenopus/genética , Xenopus laevis/metabolismo
6.
Sci Rep ; 5: 13989, 2015 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-26365306

RESUMO

Mitochondria are involved in key cellular functions including energy production, metabolic homeostasis, and apoptosis. Normal mitochondrial function is preserved by several interrelated mechanisms. One mechanism - intramitochondrial quality control (IMQC) - is represented by conserved proteases distributed across mitochondrial compartments. Many aspects and physiological roles of IMQC components remain unclear. Here, we show that the IMQC protease Oma1 is required for the stability of the respiratory supercomplexes and thus balanced and tunable bioenergetic function. Loss of Oma1 activity leads to a specific destabilization of respiratory supercomplexes and consequently to unbalanced respiration and progressive respiratory decline in yeast. Similarly, experiments in cultured Oma1-deficient mouse embryonic fibroblasts link together impeded supercomplex stability and inability to maintain proper respiration under conditions that require maximal bioenergetic output. Finally, transient knockdown of OMA1 in zebrafish leads to impeded bioenergetics and morphological defects of the heart and eyes. Together, our biochemical and genetic studies in yeast, zebrafish and mammalian cells identify a novel and conserved physiological role for Oma1 protease in fine-tuning of respiratory function. We suggest that this unexpected physiological role is important for cellular bioenergetic plasticity and may contribute to Oma1-associated disease phenotypes in humans.


Assuntos
Metaloproteases/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Animais , Linhagem Celular , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Metabolismo Energético , Larva/metabolismo , Metaloproteases/química , Metaloproteases/genética , Camundongos , Proteínas Mitocondriais/química , Proteínas Mitocondriais/genética , Morfolinos/farmacologia , Fenótipo , Estabilidade Proteica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
7.
Dev Biol ; 405(1): 108-22, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26144049

RESUMO

Folate deficiency has been associated with numerous diseases and birth defects including orofacial defects. However, whether folate has a role in the face during early orofacial development has been unclear. The present study reveals that pharmacological and antisense oligonucleotide mediated inhibition of DHFR, an integral enzyme in the folate pathway, results in specific changes in the size and shape of the midface and embryonic mouth. Such defects are accompanied by a severe reduction in the muscle and cartilage jaw elements without significant change in neural crest pattern or global levels of methylation. We propose that the orofacial defects associated with DHFR deficient function are the result of decreased cell proliferation and increased cell death via DNA damage. In particular, localized apoptosis may also be depleting the cells of the face that express crucial genes for the differentiation of the jaw structures. Folate supplementation is widely known to reduce human risk for orofacial clefts. In the present study, we show that activating folate metabolism can reduce median oral clefts in the primary palate by increasing cell survival. Moreover, we demonstrate that a minor decrease in DHFR function exacerbates median facial clefts caused by RAR inhibition. This work suggests that folate deficiencies could be a major contributing factor to multifactorial orofacial defects.


Assuntos
Fissura Palatina/embriologia , Fissura Palatina/metabolismo , Face/embriologia , Ácido Fólico/metabolismo , Boca/embriologia , Animais , Apoptose/efeitos dos fármacos , Biomarcadores/metabolismo , Cartilagem/efeitos dos fármacos , Cartilagem/embriologia , Cartilagem/patologia , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dano ao DNA , Metilação de DNA/efeitos dos fármacos , Embrião não Mamífero/anormalidades , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/patologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Leucovorina/farmacologia , Metotrexato/farmacologia , Modelos Biológicos , Morfolinos/farmacologia , Boca/metabolismo , Músculos/efeitos dos fármacos , Músculos/embriologia , Músculos/patologia , Crista Neural/efeitos dos fármacos , Crista Neural/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Receptores do Ácido Retinoico/antagonistas & inibidores , Receptores do Ácido Retinoico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tetra-Hidrofolato Desidrogenase/metabolismo , Tretinoína/metabolismo , Xenopus laevis
8.
Peptides ; 70: 17-22, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26028164

RESUMO

The enzyme ghrelin O-acyltransferase (GOAT) activates the orexigenic peptide ghrelin by transferring an acyl group from fatty acids to the serine-3 residue of the ghrelin molecule. This allows ghrelin to bind to its only known receptor, the growth hormone secretagogue receptor type 1a (GHSR1a). While studies have examined the hypothalamic transcriptional response of GOAT to metabolic challenge in mice, little has been examined in the rat hypothalamus. Furthermore, it has not been possible to identify the role of central GOAT separate from that of the periphery, since previous studies either knocked out GOAT system-wide or administered a GOAT inhibitor intraperitoneally. To determine if central GOAT expression is modulated by changes in energy state, we subjected rats to either forty-eight hours of food deprivation or three weeks of food restriction and found that GOAT mRNA increases significantly in both the hypothalamus and the stomach fundus in response to both metabolic challenges. We also found increases in hypothalamic ghrelin mRNA and stomach GHSR1a mRNA in response to food deprivation, as well as increases in hypothalamic GHSR1a mRNA in response to food restriction. We then conducted a second study where we continuously infused amorpholino antisense oligonucleotide into the lateral ventricles of rats to knock-down GOAT centrally while the animals were exposed to a high fat diet. Our results show that rats receiving the GOAT antisense gained less weight, and decreased their caloric efficiency when eating a high fat diet compared to control animals. These data suggest that central GOAT plays a role in modulating metabolism in rats.


Assuntos
Aciltransferases/antagonistas & inibidores , Peso Corporal/efeitos dos fármacos , Dieta Hiperlipídica , Mucosa Gástrica/metabolismo , Hipotálamo/metabolismo , Morfolinos/farmacologia , Aciltransferases/genética , Aciltransferases/metabolismo , Animais , Privação de Alimentos , Regulação da Expressão Gênica , Grelina/genética , Grelina/metabolismo , Hipotálamo/efeitos dos fármacos , Masculino , Ratos , Receptores de Grelina/genética , Receptores de Grelina/metabolismo
9.
Am J Pathol ; 185(4): 1061-72, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25794706

RESUMO

During cholestatic liver disease, there is dysregulation in the balance between biliary growth and loss in bile duct-ligated (BDL) rats modulated by neuroendocrine peptides via autocrine/paracrine pathways. Gonadotropin-releasing hormone (GnRH) is a trophic peptide hormone that modulates reproductive function and proliferation in many cell types. We evaluated the autocrine role of GnRH in the regulation of cholangiocyte proliferation. The expression of GnRH receptors was assessed in a normal mouse cholangiocyte cell line (NMC), sham, and BDL rats. The effect of GnRH administration was evaluated in normal rats and in NMC. GnRH-induced biliary proliferation was evaluated by changes in intrahepatic bile duct mass and the expression of proliferation and function markers. The expression and secretion of GnRH in NMC and isolated cholangiocytes was assessed. GnRH receptor subtypes GnRHR1 and GnRHR2 were expressed in cholangiocytes. Treatment with GnRH increased intrahepatic bile duct mass as well as proliferation and function markers in cholangiocytes. Transient knockdown and pharmacologic inhibition of GnRHR1 in NMC decreased proliferation. BDL cholangiocytes had increased expression of GnRH compared with normal rats, accompanied by increased GnRH secretion. In vivo and in vitro knockdown of GnRH decreased intrahepatic bile duct mass/cholangiocyte proliferation and fibrosis. GnRH secreted by cholangiocytes promotes biliary proliferation via an autocrine pathway. Disruption of GnRH/GnRHR signaling may be important for the management of cholestatic liver diseases.


Assuntos
Comunicação Autócrina , Ductos Biliares Intra-Hepáticos/citologia , Hormônio Liberador de Gonadotropina/metabolismo , Comunicação Parácrina , Animais , Ductos Biliares Intra-Hepáticos/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , AMP Cíclico/metabolismo , Imunofluorescência , Inativação Gênica/efeitos dos fármacos , Hipotálamo/metabolismo , Inositol 1,4,5-Trifosfato/metabolismo , Cirrose Hepática/patologia , Masculino , Camundongos , Morfolinos/administração & dosagem , Morfolinos/farmacologia , Comunicação Parácrina/efeitos dos fármacos , Ratos Endogâmicos F344 , Receptores LHRH/metabolismo
10.
Sci Rep ; 5: 8123, 2015 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-25630240

RESUMO

Agr family includes three groups of genes, Ag1, Agr2 and Agr3, which encode the thioredoxin domain-containing secreted proteins and have been shown recently to participate in regeneration of the amputated body appendages in amphibians. By contrast, higher vertebrates have only Agr2 and Agr3, but lack Ag1, and have low ability to regenerate the body appendages. Thus, one may hypothesize that loss of Ag1 in evolution could be an important event that led to a decline of the regenerative capacity in higher vertebrates. To test this, we have studied now the expression and role of Ag1 in the regeneration of fins of a representative of another large group of lower vertebrates, the fish Danio rerio. As a result, we have demonstrated that amputation of the Danio fins, like amputation of the body appendages in amphibians, elicits an increase of Ag1 expression in cells of the stump. Furthermore, down-regulation of DAg1 by injections of Vivo-morpholino antisense oligonucleotides resulted in a retardation of the fin regeneration. These data are in a good agreement with the assumption that the loss of Ag1 in higher vertebrates ancestors could lead to the reduction of the regenerative capacity in their modern descendants.


Assuntos
Nadadeiras de Animais/fisiologia , Isomerases de Dissulfetos de Proteínas/metabolismo , Regeneração , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Apoptose/efeitos dos fármacos , DNA Complementar/genética , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Injeções , Morfolinos/administração & dosagem , Morfolinos/farmacologia , Reação em Cadeia da Polimerase , Biossíntese de Proteínas , Isomerases de Dissulfetos de Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Regeneração/genética , Fatores de Tempo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
11.
Hum Mol Genet ; 23(17): 4651-62, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24760771

RESUMO

Duchenne muscular dystrophy (DMD) is a common and relentlessly progressive muscle disease. Some interventions have been identified that modestly slow progression and prolong survival, but more meaningful therapies are lacking. The goal of this study is to identify new therapeutic pathways for DMD using a zebrafish model of the disease. To accomplish this, we performed a non-biased drug screen in sapje, a zebrafish line with a recessive nonsense mutation in dystrophin. We identified 6 positive hits (out of 640 total drugs tested) by their ability to prevent abnormal birefringence in sapje. Follow-up analyses demonstrated that fluoxetine, a selective serotonin reuptake inhibitor (SSRI), provided the most substantial benefit. Morpholino-based experimentation confirmed that modulation of the serotonin pathway alone can prevent the dystrophic phenotype, and transcriptomic analysis revealed changes in calcium homeostasis as a potential mechanism. In all, we demonstrate that monoamine agonists can prevent disease in a vertebrate model of DMD. Given the safe and widespread use of SSRIs in clinical practice, our study identifies an attractive target pathway for therapy development.


Assuntos
Fluoxetina/uso terapêutico , Distrofia Muscular Animal/tratamento farmacológico , Distrofia Muscular de Duchenne/tratamento farmacológico , Peixe-Zebra/fisiologia , Animais , Sequência de Bases , Birrefringência , Cálcio/metabolismo , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Distrofina/metabolismo , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Azul Evans/metabolismo , Fluoxetina/farmacologia , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Homeostase/efeitos dos fármacos , Dados de Sequência Molecular , Morfolinos/farmacologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Estresse Mecânico , Análise de Sobrevida , Peixe-Zebra/embriologia , Peixe-Zebra/genética
12.
Dis Model Mech ; 6(3): 652-60, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23471908

RESUMO

Recessive mutations in KCNJ10, which encodes an inwardly rectifying potassium channel, were recently identified as the cause of EAST syndrome, a severe and disabling multi-organ disorder consisting of epilepsy, ataxia, sensorineural deafness and tubulopathy that becomes clinically apparent with seizures in infancy. A Kcnj10 knockout mouse shows postnatal mortality and is therefore not suitable for drug discovery. Because zebrafish are ideal for in vivo screening for potential therapeutics, we tested whether kcnj10 knockdown in zebrafish would fill this need. We cloned zebrafish kcnj10 and demonstrated that its function is equivalent to that of human KCNJ10. We next injected splice- and translation-blocking kcnj10 antisense morpholino oligonucleotides and reproduced the cardinal symptoms of EAST syndrome - ataxia, epilepsy and renal tubular defects. Several of these phenotypes could be assayed in an automated manner. We could rescue the morphant phenotype with complementary RNA (cRNA) encoding human wild-type KCNJ10, but not with cRNA encoding a KCNJ10 mutation identified in individuals with EAST syndrome. Our results suggest that zebrafish will be a valuable tool to screen for compounds that are potentially therapeutic for EAST syndrome or its individual symptoms. Knockdown of kcnj10 represents the first zebrafish model of a salt-losing tubulopathy, which has relevance for blood pressure control.


Assuntos
Perda Auditiva Neurossensorial/patologia , Deficiência Intelectual/patologia , Convulsões/patologia , Peixe-Zebra/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Perda Auditiva Neurossensorial/genética , Humanos , Deficiência Intelectual/genética , Rim/efeitos dos fármacos , Rim/patologia , Camundongos , Dados de Sequência Molecular , Morfolinos/farmacologia , Movimento/efeitos dos fármacos , Fenótipo , Canais de Potássio Corretores do Fluxo de Internalização/química , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Reprodutibilidade dos Testes , Convulsões/genética , Xenopus
13.
Dev Biol ; 373(2): 258-66, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23206891

RESUMO

Spontaneous intracranial hemorrhage is a debilitating form of stroke, often leading to death or permanent cognitive impairment. Many of the causative genes and the underlying mechanisms implicated in developmental cerebral-vascular malformations are unknown. Recent in vitro and in vivo studies in mice have shown inhibition of the 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) pathway to be effective in stabilizing cranial vessels. Using a combination of pharmacological and genetic approaches to specifically inhibit the HMGCR pathway in zebrafish (Danio rerio), we demonstrate a requirement for this metabolic pathway in developmental vascular stability. Here we report that inhibition of HMGCR function perturbs cerebral-vascular stability, resulting in progressive dilation of blood vessels, followed by vessel rupture, mimicking cerebral cavernous malformation (CCM)-like lesions in humans and murine models. The hemorrhages in the brain are rescued by prior exogenous supplementation with geranylgeranyl pyrophosphate (GGPP), a 20-carbon metabolite of the HMGCR pathway, required for the membrane localization and activation of Rho GTPases. Consistent with this observation, morpholino-induced depletion of the ß-subunit of geranylgeranyltransferase I (GGTase I), an enzyme that facilitates the post-translational transfer of the GGPP moiety to the C-terminus of Rho family of GTPases, mimics the cerebral hemorrhaging induced by the pharmacological and genetic ablation of HMGCR. In embryos with cerebral hemorrhage, the endothelial-specific expression of cdc42, a Rho GTPase involved in the regulation of vascular permeability, was significantly reduced. Taken together, our data reveal a metabolic contribution to the stabilization of nascent cranial vessels, requiring protein geranylgeranylation acting downstream of the HMGCR pathway.


Assuntos
Cérebro/irrigação sanguínea , Cérebro/embriologia , Hidroximetilglutaril-CoA Redutases/metabolismo , Prenilação , Transdução de Sinais , Peixe-Zebra/embriologia , Alquil e Aril Transferases/metabolismo , Animais , Atorvastatina , Hemorragia Cerebral/embriologia , Hemorragia Cerebral/patologia , Cérebro/efeitos dos fármacos , Cérebro/metabolismo , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/enzimologia , Embrião não Mamífero/patologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Células Endoteliais/patologia , Ácidos Heptanoicos/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Camundongos , Morfolinos/farmacologia , Fosfatos de Poli-Isoprenil/biossíntese , Prenilação/efeitos dos fármacos , Pirróis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
14.
Neuron ; 75(4): 688-99, 2012 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-22920259

RESUMO

Visual cues often modulate auditory signal processing, leading to improved sound detection. However, the synaptic and circuit mechanism underlying this cross-modal modulation remains poorly understood. Using larval zebrafish, we first established a cross-modal behavioral paradigm in which a preceding flash enhances sound-evoked escape behavior, which is known to be executed through auditory afferents (VIII(th) nerves) and command-like neurons (Mauthner cells). In vivo recording revealed that the visual enhancement of auditory escape is achieved by increasing sound-evoked Mauthner cell responses. This increase in Mauthner cell responses is accounted for by the increase in the signal-to-noise ratio of sound-evoked VIII(th) nerve spiking and efficacy of VIII(th) nerve-Mauthner cell synapses. Furthermore, the visual enhancement of Mauthner cell response and escape behavior requires light-responsive dopaminergic neurons in the caudal hypothalamus and D1 dopamine receptor activation. Our findings illustrate a cooperative neural mechanism for visual modulation of audiomotor processing that involves dopaminergic neuromodulation.


Assuntos
Comunicação Celular/fisiologia , Neurônios Dopaminérgicos/fisiologia , Reação de Fuga/fisiologia , Hipotálamo/citologia , Locomoção/fisiologia , Vias Visuais/fisiologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Estimulação Acústica/efeitos adversos , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Apomorfina/farmacologia , Comportamento Animal , Benzazepinas/farmacologia , Biotina/análogos & derivados , Biotina/metabolismo , Comunicação Celular/efeitos dos fármacos , Agonistas de Dopamina/farmacologia , Antagonistas de Dopamina/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Reação de Fuga/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Ácido Flufenâmico/farmacologia , Lateralidade Funcional , Ácido Glicirretínico/farmacologia , Técnicas In Vitro , Larva , Luz , Microscopia Confocal , Morfolinos/farmacologia , Técnicas de Patch-Clamp , Estimulação Luminosa/métodos , Psicoacústica , Receptores de Dopamina D1/fisiologia , Razão Sinal-Ruído , Fatores de Tempo , Valina/análogos & derivados , Valina/farmacologia , Vias Visuais/efeitos dos fármacos , Peixe-Zebra
15.
Dev Biol ; 369(1): 133-49, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22728160

RESUMO

Distinct groups of dopaminergic neurons develop at defined anatomical sites in the brain to modulate function of a large diversity of local and far-ranging circuits. However, the molecular identity as judged from transcription factor expression has not been determined for all dopaminergic groups. Here, we analyze regional expression of transcription factors in the larval zebrafish brain to determine co-expression with the Tyrosine hydroxylase marker in dopaminergic neurons. We define sets of transcription factors that clearly identify each dopaminergic group. These data confirm postulated relations to dopaminergic groups defined for mammalian systems. We focus our functional analysis on prethalamic dopaminergic neurons, which co-express the transcription factors Arx and Isl1. Morpholino-based knockdown reveals that both Arx and Isl1 are strictly required for prethalamic dopaminergic neuron development and appear to act in parallel. We further show that Arx contributes to patterning in the prethalamic region, while Isl1 is required for differentiation of prethalamic dopaminergic neurons.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Tálamo/embriologia , Fatores de Transcrição/metabolismo , Transcrição Gênica , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Animais , Padronização Corporal/efeitos dos fármacos , Padronização Corporal/genética , Catecolaminas/metabolismo , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Embrião não Mamífero/citologia , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Hipotálamo/citologia , Hipotálamo/metabolismo , Proteínas com Homeodomínio LIM/genética , Morfolinos/farmacologia , Área Pré-Óptica/citologia , Área Pré-Óptica/metabolismo , Rombencéfalo/citologia , Rombencéfalo/efeitos dos fármacos , Rombencéfalo/metabolismo , Telencéfalo/citologia , Telencéfalo/metabolismo , Tálamo/citologia , Tálamo/efeitos dos fármacos , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos , Proteínas de Peixe-Zebra/genética
16.
J Neurosci ; 32(13): 4623-31, 2012 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-22457508

RESUMO

Orexins (also called hypocretins) have been shown to be importantly involved in reward and addiction, but little is known about the circuitry that regulates orexin neuronal activity during drug-seeking behaviors. Here, we examined inputs to the lateral hypothalamus (LH) orexin cell field from the lateral septum (LS) using tract-tracing and Fos immunohistochemistry after cocaine (10 mg/kg) conditioned place preference (CPP) in Sprague Dawley rats. We found that neurons in rostral LS (LSr) that project to LH are Fos-activated in proportion to cocaine CPP, and that inhibition of LSr neurons with local baclofen and muscimol microinjection (0.3/0.03 nmol) blocks expression of Fos in LH orexin cells and cocaine preference. In addition, using local inactivation in LS and orexin antisense morpholinos in LH, we found that LSr influences on LH orexin neurons are critical for the expression of cocaine preference. These results indicate that LSr activates LH orexin neurons during cocaine place preference, and that this circuit is essential for expression of cocaine place preference.


Assuntos
Condicionamento Psicológico/fisiologia , Comportamento de Procura de Droga/fisiologia , Hipotálamo/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neuropeptídeos/fisiologia , Septo do Cérebro/fisiologia , Animais , Baclofeno/administração & dosagem , Baclofeno/farmacologia , Benzoxazóis/farmacologia , Cocaína/antagonistas & inibidores , Cocaína/farmacologia , Condicionamento Psicológico/efeitos dos fármacos , Comportamento de Procura de Droga/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Masculino , Microinjeções , Morfolinos/administração & dosagem , Morfolinos/farmacologia , Muscimol/administração & dosagem , Muscimol/farmacologia , Naftiridinas , Vias Neurais/fisiologia , Técnicas de Rastreamento Neuroanatômico/métodos , Neurônios/fisiologia , Neuropeptídeos/antagonistas & inibidores , Orexinas , Ratos , Ratos Sprague-Dawley , Septo do Cérebro/efeitos dos fármacos , Ureia/análogos & derivados , Ureia/farmacologia
17.
Clin Exp Pharmacol Physiol ; 39(1): 78-86, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21824171

RESUMO

1. Evidence is accumulating for a role for Ca²âº signalling in the differentiation and development of embryonic skeletal muscle. 2. Imaging of intact, normally developing transgenic zebrafish that express the protein component of the Ca²âº-sensitive complex aequorin, specifically in skeletal muscle, show that two distinct periods of spontaneous synchronised Ca²âº transients occur in the trunk: one at approximately 17.5-19.5 h post-fertilization (h.p.f.; termed signalling period SP1) and the other after approximately 23 h.p.f. (termed SP2). These periods of intense Ca²âº signalling activity are separated by a quiet period. 3. Higher-resolution confocal imaging of embryos loaded with the fluorescent Ca²âº reporter calcium green-1 dextran shows that the Ca²âº signals are generated almost exclusively in the slow muscle cells, the first muscle cells to differentiate, with distinct nuclear and cytoplasmic components. 4. Here, we show that coincidental with the SP1 Ca²âº signals, dystrophin becomes localized to the vertical myoseptae of the myotome. Introduction of a dmd morpholino (dmd-MO) resulted in no dystrophin being expressed in the vertical myoseptae, as well as a disruption of myotome morphology and sarcomere organization. In addition, the Ca²âº signalling signatures of dmd-MO-injected embryos or homozygous sapje mutant embryos were abnormal such that the frequency, amplitude and timing of the Ca²âº signals were altered compared with controls. 5. Our new data suggest that, in addition to a structural role, dystrophin may function in the regulation of [Ca²âº](i) during the early stages of slow muscle cell differentiation when the Ca²âº signals generated in these cells coincide with the first spontaneous contractions of the trunk.


Assuntos
Sinalização do Cálcio , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Desenvolvimento Muscular , Fibras Musculares de Contração Lenta/metabolismo , Fibras Musculares de Contração Lenta/patologia , Distrofias Musculares/metabolismo , Animais , Animais Geneticamente Modificados , Sinalização do Cálcio/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/patologia , Citoplasma/efeitos dos fármacos , Citoplasma/patologia , Desenvolvimento Embrionário/efeitos dos fármacos , Medições Luminescentes/métodos , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Microscopia Confocal , Microscopia de Fluorescência/métodos , Morfolinos/farmacologia , Contração Muscular/efeitos dos fármacos , Desenvolvimento Muscular/efeitos dos fármacos , Fibras Musculares de Contração Lenta/efeitos dos fármacos , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Distrofias Musculares/embriologia , Distrofias Musculares/patologia , Mutação , Especificidade de Órgãos , Transporte Proteico/efeitos dos fármacos , Sarcômeros/efeitos dos fármacos , Sarcômeros/metabolismo , Sarcômeros/patologia , Peixe-Zebra , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA