Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(10): e2112397119, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35239443

RESUMO

SignificanceThe modulation of growth hormone secretagogue receptor-1a (GHSR1a) signaling is a promising strategy for treating brain conditions of metabolism, aging, and addiction. GHSR1a activation results in pleiotropic physiological outcomes through distinct and pharmacologically separable G protein- and ß-arrestin (ßarr)-dependent signaling pathways. Thus, pathway-selective modulation can enable improved pharmacotherapeutics that can promote therapeutic efficacy while mitigating side effects. Here, we describe the discovery of a brain-penetrant small molecule, N8279 (NCATS-SM8864), that biases GHSR1a conformations toward Gαq activation and reduces aberrant dopaminergic behavior in mice. N8279 represents a promising chemical scaffold to advance the development of better treatments for GHSR1a-related brain disorders involving the pathological dysregulation of dopamine.


Assuntos
Encéfalo/metabolismo , Dopamina/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Receptores de Grelina/metabolismo , Animais , Dopamina/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Masculino , Camundongos , Camundongos Knockout , Receptores de Grelina/genética
2.
Genes Brain Behav ; 16(5): 522-536, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28239983

RESUMO

Parkinson's disease is a debilitating neurodegenerative condition for which there is no cure. Converging evidence implicates gangliosides in the pathogenesis of several neurodegenerative diseases, suggesting a potential new class of therapeutic targets. We have shown that interventions that simultaneously increase the neuroprotective GM1 ganglioside and decrease the pro-apoptotic GD3 ganglioside - such as inhibition of GD3 synthase (GD3S) or administration of sialidase - are neuroprotective in vitro and in a number of preclinical models. In this study, we investigated the effects of GD3S deletion on parkinsonism induced by 1-methyl-4phenyl-1,2,3,6-tetrahydropyridine (MPTP). MPTP was administered to GD3S-/- mice or controls using a subchronic regimen consisting of three series of low-dose injections (11 mg/kg/day × 5 days each, 3 weeks apart), and motor function was assessed after each. The typical battery of tests used to assess parkinsonism failed to detect deficits in MPTP-treated mice. More sensitive measures - such as the force-plate actimeter and treadmill gait parameters - detected subtle effects of MPTP, some of which were absent in mice lacking GD3S. In wild-type mice, MPTP destroyed 53% of the tyrosine-hydroxylase (TH)-positive neurons in the substantia nigra pars compacta (SNc) and reduced striatal dopamine 60.7%. In contrast, lesion size was only 22.5% in GD3S-/- mice and striatal dopamine was reduced by 37.2%. Stereological counts of Nissl-positive SNc neurons that did not express TH suggest that neuroprotection was complete but TH expression was suppressed in some cells. These results show that inhibition of GD3S has neuroprotective properties in the MPTP model and may warrant further investigation as a therapeutic target.


Assuntos
Deleção de Genes , Intoxicação por MPTP/genética , Sialiltransferases/genética , Animais , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Locomoção , Camundongos , Camundongos Endogâmicos C57BL , Substância Negra/metabolismo , Substância Negra/patologia , Substância Negra/fisiopatologia
3.
Mol Psychiatry ; 20(6): 744-54, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25330741

RESUMO

Major depressive disorder is often linked to stress. Although short-term stress is without effect in mice, prolonged stress leads to depressive-like behavior, indicating that an allostatic mechanism exists in this difference. Here we demonstrate that mice after short-term (1 h per day for 7 days) chronic restraint stress (CRS), do not display depressive-like behavior. Analysis of the hippocampus of these mice showed increased levels of neurotrophic factor-α1 (NF-α1; also known as carboxypeptidase E, CPE), concomitant with enhanced fibroblast growth factor 2 (FGF2) expression, and an increase in neurogenesis in the dentate gyrus. In contrast, after prolonged (6 h per day for 21 days) CRS, mice show decreased hippocampal NF-α1 and FGF2 levels and depressive-like responses. In NF-α1-knockout mice, hippocampal FGF2 levels and neurogenesis are reduced. These mice exhibit depressive-like behavior that is reversed by FGF2 administration. Indeed, studies in cultured hippocampal neurons reveal that NF-α1 treatment directly upregulates FGF2 expression through extracellular signal-regulated kinase-Sp1 signaling. Thus, during short-term CRS, hippocampal NF-α1 expression is upregulated and has a key role in preventing the onset of depressive-like behavior through enhanced FGF2-mediated neurogenesis. To evaluate the therapeutic potential of this pathway, we examined, rosiglitazone (Rosi), a PPARγ agonist, which has been shown to have antidepressant activity in rodents and humans. Rosi upregulates FGF2 expression in a NF-α1-dependent manner in hippocampal neurons. Mice fed Rosi show increased hippocampal NF-α1 levels and neurogenesis compared with controls, thereby indicating the antidepressant action of this drug. Development of drugs that activate the NF-α1/FGF2/neurogenesis pathway can offer a new approach to depression therapy.


Assuntos
Carboxipeptidase H/metabolismo , Depressão/prevenção & controle , Hipocampo/citologia , Hipoglicemiantes/uso terapêutico , Neurogênese/efeitos dos fármacos , Tiazolidinedionas/uso terapêutico , Animais , Carboxipeptidase H/genética , Células Cultivadas , Depressão/etiologia , Depressão/genética , Modelos Animais de Doenças , Proteínas do Domínio Duplacortina , Preferências Alimentares/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Neuropeptídeos/metabolismo , Rosiglitazona , Estresse Psicológico/complicações , Sacarose/administração & dosagem , Edulcorantes , Natação/psicologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
4.
J Neurophysiol ; 111(10): 1927-39, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24554785

RESUMO

Mice with a single copy of the peptide amidating monooxygenase (Pam) gene (PAM(+/-)) are impaired in contextual and cued fear conditioning. These abnormalities coincide with deficient long-term potentiation (LTP) at excitatory thalamic afferent synapses onto pyramidal neurons in the lateral amygdala. Slice recordings from PAM(+/-) mice identified an increase in GABAergic tone (Gaier ED, Rodriguiz RM, Ma XM, Sivaramakrishnan S, Bousquet-Moore D, Wetsel WC, Eipper BA, Mains RE. J Neurosci 30: 13656-13669, 2010). Biochemical data indicate a tissue-specific deficit in Cu content in the amygdala; amygdalar expression of Atox-1 and Atp7a, essential for transport of Cu into the secretory pathway, is reduced in PAM(+/-) mice. When PAM(+/-) mice were fed a diet supplemented with Cu, the impairments in fear conditioning were reversed, and LTP was normalized in amygdala slice recordings. A role for endogenous Cu in amygdalar LTP was established by the inhibitory effect of a brief incubation of wild-type slices with bathocuproine disulfonate, a highly selective, cell-impermeant Cu chelator. Interestingly, bath-applied CuSO4 had no effect on excitatory currents but reversibly potentiated the disynaptic inhibitory current. Bath-applied CuSO4 was sufficient to potentiate wild-type amygdala afferent synapses. The ability of dietary Cu to affect signaling in pathways that govern fear-based behaviors supports an essential physiological role for Cu in amygdalar function at both the synaptic and behavioral levels. This work is relevant to neurological and psychiatric disorders in which disturbed Cu homeostasis could contribute to altered synaptic transmission, including Wilson's, Menkes, Alzheimer's, and prion-related diseases.


Assuntos
Tonsila do Cerebelo/fisiologia , Cobre/metabolismo , Animais , Condicionamento Psicológico/fisiologia , Cobre/administração & dosagem , Dieta , Medo/fisiologia , Feminino , Hipocampo/fisiologia , Potenciação de Longa Duração/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Inibição Neural/fisiologia , Plasticidade Neuronal/fisiologia , Técnicas de Patch-Clamp , Células Piramidais/fisiologia , Sinapses/fisiologia , Transmissão Sináptica , Tálamo/fisiologia , Técnicas de Cultura de Tecidos
5.
Genes Brain Behav ; 9(3): 257-68, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20050925

RESUMO

Synapsin III is a neuron-specific phosphoprotein that plays an important role in synaptic transmission and neural development. While synapsin III is abundant in embryonic brain, expression of the protein in adults is reduced and limited primarily to the hippocampus, olfactory bulb and cerebral cortex. Given the specificity of synapsin III to these brain areas and because it plays a role in neurogenesis in the dentate gyrus, we investigated whether it may affect learning and memory processes in mice. To address this point, synapsin III knockout mice were examined in a general behavioral screen, several tests to assess learning and memory function, and conditioned fear. Mutant animals displayed no anomalies in sensory and motor function or in anxiety- and depressive-like behaviors. Although mutants showed minor alterations in the Morris water maze, they were deficient in object recognition 24 h and 10 days after training and in social transmission of food preference at 20 min and 24 h. In addition, mutants displayed abnormal responses in contextual and cued fear conditioning when tested 1 or 24 h after conditioning. The synapsin III knockout mice also showed aberrant responses in fear-potentiated startle. As synapsin III protein is decreased in schizophrenic brain and because the mutant mice do not harbor obvious anatomical deficits or neurological disorders, these mutants may represent a unique neurodevelopmental model for dissecting the molecular pathways that are related to certain aspects of schizophrenia and related disorders.


Assuntos
Condicionamento Psicológico/fisiologia , Medo , Rememoração Mental/fisiologia , Reconhecimento Psicológico/fisiologia , Sinapsinas/genética , Análise de Variância , Animais , Aprendizagem por Associação/fisiologia , Comportamento Animal/fisiologia , Preferências Alimentares/fisiologia , Hipocampo/metabolismo , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Knockout , Atividade Motora/genética , Neurônios/metabolismo , Reflexo de Sobressalto/genética , Comportamento Social , Comportamento Espacial/fisiologia , Sinapsinas/metabolismo
6.
Genes Brain Behav ; 7(4): 418-26, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21450041

RESUMO

Endothelin-converting enzyme (ECE)-2 is a metalloprotease that possesses many properties consistent with it being a neuropeptide-processing enzyme. This protease is found primarily in neural tissues, with high levels of expression in midbrain, cerebellum, hypothalamus, frontal cortex and spinal cord and moderate levels in hippocampus and striatum. To evaluate its role in neural function, mice have been generated lacking this enzyme. Physical appearance, autonomic reflexes, motor co-ordination, balance, locomotor activity and spontaneous emotional responses appear normal in these knockout (KO) mice. However, these mutants display deficits in learning and memory as evidenced by marked impairment in the Morris water maze. Knockout mice are also deficient in object recognition memory where they show delays in discerning changes in object location and in recognizing the introduction of a novel object. In this study, perseveration appears to interfere with learning and memory. Finally, mutants are impaired in social transmission of food preference where they show poor short-term memory and perturbations in long-term memory; the latter can be ameliorated by reminder cues. As ECE-2 has been implicated in Alzheimer's disease, the deficits in learning and memory in the KO mice may provide unique insights into processes that may contribute to this disease and possible other disorders of cognition.


Assuntos
Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/fisiologia , Deficiências da Aprendizagem/genética , Deficiências da Aprendizagem/psicologia , Transtornos da Memória/genética , Transtornos da Memória/psicologia , Metaloendopeptidases/genética , Metaloendopeptidases/fisiologia , Animais , Ácido Aspártico Endopeptidases/deficiência , Comportamento Animal/fisiologia , Emoções/fisiologia , Enzimas Conversoras de Endotelina , Comportamento Exploratório/fisiologia , Feminino , Preferências Alimentares/fisiologia , Masculino , Aprendizagem em Labirinto/fisiologia , Metaloendopeptidases/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia , Neuropeptídeos/metabolismo , Sistemas Neurossecretores/fisiologia , RNA/genética , RNA/isolamento & purificação , Reconhecimento Psicológico/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Filtro Sensorial/genética , Filtro Sensorial/fisiologia , Comportamento Social
7.
Neuroscience ; 132(4): 1055-72, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15857710

RESUMO

Several lines of research have implicated glutathione (GSH) in schizophrenia. For instance, GSH deficiency has been reported in the prefrontal cortex of schizophrenics in vivo. Further, in rats postnatal GSH-deficiency combined with hyperdopaminergia led to cognitive impairments in the adult. In the present report we studied the effects of 2-day GSH-deficiency with L-buthionine-(S,R)-sulfoximine on monoaminergic function in mice. The effect of GSH-deficiency per se and when combined with the amphetamine and phencyclidine (PCP) models of schizophrenia was investigated. GSH-deficiency significantly altered tissue levels of dopamine (DA), 5-hydroxytryptamine (5-HT) and their respective metabolites homovanillic acid (HVA), and 5-hydroxyindoleacetic acid (5-HIAA) in a region-specific fashion. The effects of GSH-deficiency on tissue monoamines were distinct from and, generally, did not interact with the effects of amphetamine (5 mg/kg; i.p.) on tissue monoamines. Microdialysis studies showed that extracellular DA-release after amphetamine (5 mg/kg, i.p.) was two-fold increased in the nucleus accumbens of GSH-deficient mice as compared with control mice. Basal DA was unaltered. Further, extracellular levels of HVA in the frontal cortex and hippocampus and 5-HIAA in the nucleus accumbens were elevated by GSH-deficiency per se. Spontaneous locomotor activity in the open field was unchanged in GSH-deficient mice. In contrast, GSH-deficiency modulated the locomotor responses to mid-range doses of amphetamine (1.5 and 5 mg/kg, i.p.). Further, GSH-deficient mice displayed an increased locomotor response to low (2 and 3 mg/kg, i.p.) doses of phencyclidine (PCP). In conclusion, the data presented here show that even short-term GSH-deficiency has consequences for DA and 5-HT function. This was confirmed on both neurochemical and behavioral levels. How GSH and the monoamines interact needs further scrutiny. Moreover, the open field findings suggest reduced or altered N-methyl-d-aspartate (NMDA) receptor function in GSH-deficient mice. Thus, GSH-deficiency can lead to disturbances in DA, 5-HT and NMDA function, a finding that may have relevance for schizophrenia.


Assuntos
Dopamina/metabolismo , Glutationa/deficiência , Esquizofrenia/fisiopatologia , Serotonina/metabolismo , Anfetamina/toxicidade , Animais , Encéfalo/metabolismo , Cromatografia Líquida de Alta Pressão , Dopamina/análise , Alucinógenos/toxicidade , Ácido Homovanílico/metabolismo , Ácido Hidroxi-Indolacético/metabolismo , Masculino , Camundongos , Microdiálise , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Fenciclidina/toxicidade , Receptores de N-Metil-D-Aspartato/metabolismo , Esquizofrenia/induzido quimicamente , Serotonina/análise
8.
Neuroscience ; 118(2): 297-310, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12699766

RESUMO

Dopamine (DA) is a neurotransmitter that has been implicated in a wide variety of psychiatric disorders that include attention deficit-hyperactivity disorder (ADHD), schizophrenia, and drug abuse. Recently, we have been working with a mouse in which the gene for the DA transporter (DAT) has been disrupted. This mouse is hyperactive in the open field, displays an inability to inhibit ongoing behaviors, and is deficient on learning and memory tasks. Psychostimulants such as amphetamine and methylphenidate attenuate the hyperlocomotion of the mutants, but stimulate activity of the wild type (WT) controls. The objective of the present study is to examine the neural basis for the differential responses to psychostimulants in these mice. WT and DAT knockout (KO) animals were given vehicle or methylphenidate, amphetamine, or cocaine and brain sections were immunostained for Fos. In WT mice, methylphenidate induced Fos-like immunoreactivity (Fos-LI) in the mesostriatal and mesolimbocortical DA pathways that included the anterior olfactory nucleus, frontal association cortex, orbitofrontal cortex, cingulate cortex, caudate-putamen, globus pallidus, claustrum, lateral septum, nucleus accumbens, basolateral and central nuclei of the amygdala, bed nucleus of stria terminalis, subthalamic nucleus, substantia nigra, ventral tegmental area, and dorsal raphe. Additional areas of activation included the granular dentate gyrus, Edinger-Westphal nucleus, and periaqueductal gray. While the mutants showed little response in most of these same areas, the anterior olfactory nucleus, caudal caudate-putamen, lateral septum, basolateral and central nuclei of the amygdala, and bed nucleus of stria terminalis were activated. Amphetamine and cocaine produced similar changes to that for methylphenidate, except these psychostimulants also induced Fos-LI in the nucleus accumbens of the KO animals. Since the DAT gene is disrupted in the KO mouse, these findings suggest that dopaminergic mechanisms may mediate the WT responses, whereas non-dopaminergic systems predominate in the mutant. In the mutants, it appears that limbic areas and non-dopaminergic transmitter systems within these brain regions may mediate responses to psychostimulants. Inasmuch as the KO mouse may represent a useful animal model for ADHD and because psychostimulants such as cocaine are reinforcing to these animals, our results may provide some useful insights into the neural mechanisms-other than DA-that may contribute to the symptoms of ADHD and/or drug abuse in human patients.


Assuntos
Estimulantes do Sistema Nervoso Central/farmacologia , Glicoproteínas de Membrana , Proteínas de Membrana Transportadoras/metabolismo , Proteínas do Tecido Nervoso , Vias Neurais/efeitos dos fármacos , Anfetamina/farmacologia , Animais , Western Blotting , Contagem de Células , Proteínas da Membrana Plasmática de Transporte de Dopamina , Feminino , Imuno-Histoquímica , Masculino , Proteínas de Membrana Transportadoras/genética , Metilfenidato/farmacologia , Camundongos , Camundongos Knockout , Mutação , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fatores de Tempo
9.
Endocrinology ; 142(11): 4839-51, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11606452

RESUMO

Norepinephrine has long been known to stimulate the pulsatile and preovulatory release of LH-releasing hormone (LHRH). In vivo and in vitro studies indicate that these effects are mediated primarily through alpha(1)-adrenergic receptors (alpha(1)-ARs). With the immortalized hypothalamic LHRH neurons, we have found that alpha(1)-adrenergic agents directly stimulate the secretion of LHRH in a dose-dependent manner. Ligand binding and RNA studies demonstrate that the GT1 cells contain both alpha(1A)- and alpha(1B)-ARs. Competition binding experiments show that approximately 75% of the binding is due to alpha(1B)-ARs; the remainder is made up of alpha(1A)-ARs. Receptor activation leads to stimulation of PLC. PLC beta 1 and PLC beta 3 are expressed in GT1 neurons, and these PLCs are probably responsible for the release of diacylglycerol and IP as well as the increase in intracellular calcium. The mobilization of cytoplasmic calcium is sufficient to stimulate cytosolic PLA(2) (cPLA(2)) and release arachidonic acid. A dissection of the contributions of the phospholipases to LHRH secretion suggests that cPLA(2) acts downstream of PLC and that it significantly augments the PLC-stimulated LHRH secretory response. Inasmuch as the alpha(1)-ARs are known to play a critical role in LHRH physiology, we propose that both PLC and cPLA(2) are critical in regulating and amplifying LHRH release.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/fisiologia , Neurônios/fisiologia , Fosfolipases A/fisiologia , Receptores Adrenérgicos alfa/fisiologia , Fosfolipases Tipo C/fisiologia , Ácido Araquidônico/metabolismo , Cálcio/metabolismo , Linhagem Celular Transformada , Citoplasma/metabolismo , Citosol/enzimologia , Hipotálamo/citologia , Ligantes , Neurônios/citologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , RNA Mensageiro/metabolismo , Receptores Adrenérgicos alfa/genética
10.
J Neurosci ; 20(22): RC107, 2000 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11069976

RESUMO

The Ca(2+)/calmodulin-dependent protein kinase CaMKIV was first identified in the cerebellum and has been implicated in nuclear signaling events that control neuronal growth, differentiation, and plasticity. To understand the physiological importance of CaMKIV, we disrupted the mouse Camk4 gene. The CaMKIV null mice displayed locomotor defects consistent with altered cerebellar function. Although the overall cytoarchitecture of the cerebellum appeared normal in the Camk4(-/-) mice, we observed a significant reduction in the number of mature Purkinje neurons and reduced expression of the protein marker calbindin D28k within individual Purkinje neurons. Western immunoblot analyses of cerebellar extracts also established significant deficits in the phosphorylation of cAMP response element-binding protein at serine-133, a proposed target of CaMKIV. Additionally, the absence of CaMKIV markedly altered neurotransmission at excitatory synapses in Purkinje cells. Multiple innervation by climbing fibers and enhanced parallel fiber synaptic currents suggested an immature development of Purkinje cells in the Camk4(-/-) mice. Together, these findings demonstrate that CaMKIV plays key roles in the function and development of the cerebellum.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/deficiência , Doenças Cerebelares/enzimologia , Doenças Cerebelares/genética , Cerebelo/enzimologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/farmacocinética , Animais , Comportamento Animal , Western Blotting , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteína Quinase Tipo 4 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Doenças Cerebelares/fisiopatologia , Cerebelo/patologia , Cerebelo/fisiopatologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/genética , Heterozigoto , Homozigoto , Técnicas In Vitro , Camundongos , Camundongos Knockout , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Células de Purkinje/enzimologia , Células de Purkinje/patologia
11.
Brain Res Mol Brain Res ; 79(1-2): 18-31, 2000 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-10925140

RESUMO

Glia in the brain respond to various toxins with an increased expression of inducible nitric oxide synthase (iNOS) and an increased production of nitric oxide (NO). Here, we report that lipopolysaccharide (LPS)-induced expression of iNOS was down-regulated post-transcriptionally through the destabilization of iNOS mRNA by the indolocarbazole compound, Gö6976, in murine microglia. This Gö6976 effect is specific for iNOS since tumor necrosis factor alpha was unaffected by the compound. Interestingly, the post-transcriptional effects ascribed to Gö6976 were not observed with other inhibitors of protein kinase A, C (PKC), G, or protein tyrosine kinases. Instead, these kinases appear to affect the iNOS/NO system at the transcriptional level. In the past, Gö6976 has been reported to be a rather specific inhibitor of PKC in vitro. Results from our experiments, through prolonged treatment with phorbol esters and with the various PKC inhibitors including phorbol ester-insensitive PKC isotype inhibitor, suggest that the Gö6976-mediated post-transcriptional regulation of iNOS gene expression and NO production in microglia is not mediated through its reputed effects on PKC activity. Since the effects of various neurotoxins and certain neurodegenerative diseases may be manifested through alterations in the iNOS/NO system, post-transcriptional control of this system may represent a novel strategy for therapeutic intervention.


Assuntos
Carbazóis/farmacologia , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica , Indóis/farmacologia , Lipopolissacarídeos/farmacologia , Neuroglia/enzimologia , Óxido Nítrico Sintase/genética , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , Animais , Encéfalo/citologia , Encéfalo/enzimologia , Células Cultivadas , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Cinética , Camundongos , Neuroglia/citologia , Óxido Nítrico Sintase Tipo II , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Acetato de Tetradecanoilforbol/farmacologia , Transcrição Gênica/efeitos dos fármacos
12.
Brain Res Mol Brain Res ; 79(1-2): 32-44, 2000 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-10925141

RESUMO

The expression of inducible nitric oxide synthase (iNOS) and the production of nitric oxide (NO) after exposure to endotoxins has been implicated in immune-mediated neurotoxicity. The indolocarbazole compound Gö6976, which has been described as a selective protein kinase C (PKC) inhibitor in vitro, rescued neurons from lipopolysaccharide/interferon-gamma (LPS/IFNgamma)- or interleukin-1alpha/tumor necrosis alpha/IFNgamma (IL-1alpha/TNFalpha/IFNgamma)-induced cytotoxicity in murine primary neuron-glia co-cultures. Other compounds known to inhibit PKC, Ro31-8220, GF109203X, Gö7874, H7, staurosporine and H89, failed to rescue neurons from the LPS/IFNgamma-induced cytotoxicity. These results suggest that the neuroprotection by Gö6976 from the LPS/IFNgamma-induced neuronal cell death is not mediated through its reputed effects on PKC activity. The neuroprotection paralleled the inhibition of iNOS gene expression and NO production. However, further analyses correlating NO production with the extent of neurotoxicity suggested that additional mechanism(s) besides the inhibition of the iNOS/NO system may be responsible for the neuroprotective effects of Gö6976. An understanding of the mechanism underlying the neuroprotective effect of Gö6976 may provide key insights into potential interventions for immune-mediated neurodegenerative diseases.


Assuntos
Carbazóis/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Indóis/farmacologia , Interferon gama/toxicidade , Lipopolissacarídeos/toxicidade , Neuroglia/citologia , Neurônios/citologia , Fármacos Neuroprotetores/farmacologia , Animais , Animais Recém-Nascidos , Técnicas de Cocultura , Regulação da Expressão Gênica , Interleucina-1/farmacologia , Camundongos , Proteínas Associadas aos Microtúbulos/análise , Moléculas de Adesão de Célula Nervosa/análise , Neuroglia/efeitos dos fármacos , Neuroglia/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Proteína Quinase C/antagonistas & inibidores , Proteínas Recombinantes/toxicidade , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Acetato de Tetradecanoilforbol/farmacologia , Fator de Necrose Tumoral alfa/toxicidade
13.
Nat Neurosci ; 3(5): 465-71, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10769386

RESUMO

The action of norepinephrine (NE) is terminated, in part, by its uptake into presynaptic noradrenergic neurons by the plasma-membrane NE transporter (NET), which is a target for antidepressants and psychostimulants. Disruption of the NET gene in mice prolonged the clearance of NE and elevated extracellular levels of this catecholamine. In a classical test for antidepressant drugs, the NET-deficient (NET-/-) animals behaved like antidepressant-treated wild-type mice. Mutants were hyper-responsive to locomotor stimulation by cocaine or amphetamine. These responses were accompanied by dopamine D2/D3 receptor supersensitivity. Thus altering NET expression significantly modulates midbrain dopaminergic function, an effect that may be an important component of the actions of antidepressants and psychostimulants.


Assuntos
Antidepressivos/farmacologia , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Estimulantes do Sistema Nervoso Central/farmacologia , Deleção de Genes , Simportadores , Anfetamina/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Cocaína/farmacologia , Dopamina/metabolismo , Dopamina/farmacologia , Agonistas de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Regulação para Baixo , Homeostase , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Norepinefrina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Norepinefrina , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D3 , Transmissão Sináptica/efeitos dos fármacos
14.
Eur J Biochem ; 262(2): 569-74, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10336644

RESUMO

Endoproteolytic cleavage of protein prohormones often generates intermediates extended at the C-terminus by Arg-Arg or Lys-Arg, the removal of which by a carboxypeptidase (CPE) is normally an important step in the maturation of many peptide hormones. Recent studies in mice that lack CP activity indicate the existence of alternative tissue or plasma enzymes capable of removing C-terminal basic residues from prohormone intermediates. Using inhibitors of angiotensin I-converting enzyme (ACE) and CP, we show that both these enzymes in mouse serum can remove the basic amino acids from the C-terminus of CCK5-GRR and LH-RH-GKR, but only CP is responsible for converting diarginyl insulin to insulin. ACE activity removes C-terminal dipeptides to generate the Gly-extended peptides, whereas CP hydrolysis gives rise to CCK5-GR and LH-RH-GK, both of which are susceptible to the dipeptidyl carboxypeptidase activity of ACE. Somatic ACE has two similar protein domains (the N-domain and the C-domain), each with an active site that can display different substrate specificities. CCK5-GRR is a high-affinity substrate for both the N-domain and C-domain active sites of human sACE (Km of 9.4 microm and 9.0 microm, respectively) with the N-domain showing greater efficiency (kcat : Km ratio of 2.6 in favour of the N-domain). We conclude that somatic forms of ACE should be considered as alternatives to CPs for the removal of basic residues from some Arg/Lys-extended peptides.


Assuntos
Colecistocinina/química , Dipeptídeos/metabolismo , Gastrinas/química , Hormônio Liberador de Gonadotropina/química , Insulina/análogos & derivados , Peptidil Dipeptidase A/metabolismo , Animais , Arginina/química , Feminino , Glicina/química , Humanos , Hidrólise , Insulina/química , Lisina/química , Camundongos , Especificidade por Substrato
15.
Endocrinology ; 140(6): 2685-95, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10342859

RESUMO

Mice carrying a null mutation for either of the two cyclooxygenase (COX) isoenzymes, necessary for prostanoid production, exhibit several isotype-specific reproductive abnormalities. Mice deficient in COX-1 are fertile but have decreased pup viability, whereas mice deficient in COX-2 fail to ovulate and have abnormal implantation and decidualization responses. The present study identifies the specific contribution of each COX isoenzyme in hypothalamic, pituitary, and ovarian function and establishes the pathology and rescue of the anovulatory syndrome in the COX-2-deficient mouse. In both COX-1- and COX-2-deficient mice, pituitary gonadotropins were selectively increased, whereas hypothalamic LHRH and serum gonadotropin levels were similar to those in wild-type animals (+/+). No significant differences in serum estrogen or progesterone were noted among the three genotypes. Exogenous gonadotropin stimulation with PMSG and hCG produced a comparable 4-fold increase in ovarian PGE2 levels in wild-type and COX-1(-/-) mice. COX-2(-/-) mice had no increase in PGE2 over PMSG-stimulated levels. Wild-type and COX-1(-/-) mice ovulated in response to PMSG/hCG; very few COX-2(-/-) animals responded to this regimen. The defect in ovulation in COX-2 mutants was attributed to both an abnormal cumulus oophorum expansion and subsequent stigmata formation. Gonadotropin stimulation and concurrent treatment with PGE2 or interleukin-1beta resulted in ovulation of COX-2(-/-) mice comparable to that in COX-2(+/+), whereas treatment with PGF2alpha was less effective. Collectively, these data demonstrate that COX-2, but not COX-1, is required for the gonadotropin induction of ovarian PG levels; that COX-2-related prostanoids are required for stabilization of the cumulus oophorum during ovulation; and that ovulation can be restored in the COX-2(-/-) animals by simultaneous treatment with gonadotropins and PGE2 or interleukin-1beta.


Assuntos
Anovulação/tratamento farmacológico , Dinoprostona/farmacologia , Interleucina-1/farmacologia , Isoenzimas/fisiologia , Prostaglandina-Endoperóxido Sintases/fisiologia , Animais , Anovulação/etiologia , Gonadotropina Coriônica/farmacologia , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Dinoprostona/análise , Estro , Feminino , Fertilidade , Hormônio Foliculoestimulante/análise , Isoenzimas/deficiência , Proteínas de Membrana , Camundongos , Ovulação/efeitos dos fármacos , Prostaglandina-Endoperóxido Sintases/deficiência
16.
Science ; 283(5400): 397-401, 1999 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9888856

RESUMO

The mechanism by which psychostimulants act as calming agents in humans with attention-deficit hyperactivity disorder (ADHD) or hyperkinetic disorder is currently unknown. Mice lacking the gene encoding the plasma membrane dopamine transporter (DAT) have elevated dopaminergic tone and are hyperactive. This activity was exacerbated by exposure to a novel environment. Additionally, these mice were impaired in spatial cognitive function, and they showed a decrease in locomotion in response to psychostimulants. This paradoxical calming effect of psychostimulants depended on serotonergic neurotransmission. The parallels between the DAT knockout mice and individuals with ADHD suggest that common mechanisms may underlie some of their behaviors and responses to psychostimulants.


Assuntos
Estimulantes do Sistema Nervoso Central/farmacologia , Corpo Estriado/metabolismo , Hipercinese/tratamento farmacológico , Proteínas de Membrana Transportadoras , Proteínas do Tecido Nervoso , Serotonina/fisiologia , Simportadores , Transmissão Sináptica , Animais , Transtorno do Deficit de Atenção com Hiperatividade/tratamento farmacológico , Transtorno do Deficit de Atenção com Hiperatividade/fisiopatologia , Transtorno do Deficit de Atenção com Hiperatividade/psicologia , Comportamento Animal/efeitos dos fármacos , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Dopamina/metabolismo , Dopamina/fisiologia , Proteínas da Membrana Plasmática de Transporte de Dopamina , Fluoxetina/farmacologia , Humanos , Hipercinese/fisiopatologia , Hipercinese/psicologia , Aprendizagem em Labirinto , Glicoproteínas de Membrana/efeitos dos fármacos , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Proteínas da Membrana Plasmática de Transporte de Norepinefrina , Serotonina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina , Inibidores Seletivos de Recaptação de Serotonina/farmacologia
17.
Endocrinology ; 139(11): 4598-606, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9794471

RESUMO

We have recently shown that protein kinase C (PKC) modifies estrogen receptor (ER) binding and modulates the responsiveness to estrogens in a clonal osteoblast-like cell line stably transfected with the ER. The purpose of the present study was to determine whether the interaction observed between the ER and PKC signaling in these cells occurs in additional estrogen target organs, such as the uterus. When uteri were incubated for 2 h with increasing concentrations of a kinase inhibitor (H7), ER binding was enhanced in a dose-dependent manner. Stimulation of PKC with phorbol ester reduced PKC activity levels, but increased ER binding. Interestingly, the changes in binding appeared to be due primarily to alterations in cytosolic ER levels, as binding in the nuclear fraction was minimally enhanced. When levels of ER messenger RNA were evaluated by Northern blot analysis, no differences were observed among the H7- or 12-O-tetradecanoylphorbol-13-acetate (TPA)-treated and untreated groups. Western blot analysis, however, demonstrated that levels of ER cytosolic protein in the H7-, TPA-, and staurosporine-treated groups were increased relative to those in the untreated controls. When uteri were incubated with diethylstilbestrol in the presence of either H7 or TPA, no change in cytosolic ER levels was found, suggesting that only unoccupied ERs are responsive to modulation by PKC. Western blotting of the various PKC isoforms indicated that although PKC alpha, -beta1, -betaII, -delta, and -zeta are expressed in the uterus, only PKC alpha and -beta1 are translocated from the soluble to the particulate fraction and then degraded after phorbol ester stimulation. Hence, one or both of these latter PKC isoforms may regulate cytosolic ER levels. Collectively, these data indicate that PKC may play an important role in the modulation of uterine ER levels and that PKC may exert its effect on the ER at some posttranscriptional or posttranslational step. Finally, our results show that an ER-PKC interaction occurs in a whole organ such as the uterus and that this interaction may be important in the regulation of the ER activity in a variety of estrogen-responsive tissues.


Assuntos
Isoenzimas/fisiologia , Proteína Quinase C/fisiologia , Receptores de Estrogênio/metabolismo , Útero/enzimologia , Útero/metabolismo , Animais , Northern Blotting , Western Blotting , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Feminino , Técnicas In Vitro , Isoenzimas/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos ICR , Osteoblastos/enzimologia , Osteoblastos/metabolismo , Ovariectomia , Proteína Quinase C/antagonistas & inibidores , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
18.
Endocrinology ; 139(10): 4092-101, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9751487

RESUMO

Testicular androgens are integral components of the hormonal feedback loops that regulate circulating levels of LHbeta and FSH. The sites of feedback include hypothalamic areas regulating GnRH neurons and pituitary gonadotropes. To better define the roles of androgen receptor (AR), estrogen receptor-alpha (ERalpha), and estrogen receptor-beta (ERbeta) in mediating feedback effects of sex steroids on reproductive neuroendocrine function, we have determined the effects of castration and steroid replacement therapy on hypothalamic GnRH content, pituitary LHbeta and FSHbeta messenger RNA (mRNA) levels, and serum gonadotropins in male wild-type (WT) and estrogen receptor-alpha knockout (ERKO) mice. Hypothalami from intact WT and ERKO males contained similar amounts of GnRH, whereas castration significantly reduced GnRH contents in both genotypes. Replacement therapy with estradiol (E2), testosterone (T), or dihydrotestosterone (DHT) restored hypothalamic GnRH content in castrated (CAST) WT mice; only the androgens were effective in CAST ERKOs. Analyses of pituitary function revealed that LHbeta mRNA and serum LHbeta levels in intact ERKOs were 2-fold higher than those in intact WT males. Castration increased levels of LHbeta mRNA (1.5- to 2-fold) and serum LHbeta (4- to 5-fold) in both genotypes. Both E2 and T treatments significantly suppressed LHbeta mRNA and serum LH levels in CAST WT males. However, E2 was completely ineffective, and T was only partially effective in suppressing these two indexes in the CAST ERKO males. DHT treatments stimulated a 50% increase in LHbeta mRNA and serum LH levels in WT males, whereas serum LH was significantly suppressed in DHT-treated ERKO males. Although the pituitaries from intact ERKO males contained similar amounts of FSHbeta mRNA, serum FSH levels were 20% higher than those in the intact WT males. Castration increased FSHbeta mRNA levels only in WT males, but significantly increased serum FSH levels in both genotypes. Both E2 and T treatments significantly suppressed serum FSH in CAST WT males, whereas only E2 suppressed FSHbeta mRNA. DHT treatments of CAST WT mice stimulated a small increase in serum FSH, but failed to alter FSHbeta mRNA levels. None of the steroid treatments exerted any significant effect on FSHbeta mRNA or serum FSH levels in CAST ERKOs. These data suggest that hypothalamic GnRH contents can be maintained solely through AR signaling pathways. However, normal regulation of gonadotrope function requires aromatization of T and activation of ERalpha signaling pathways in the gonadotrope. In addition, serum FSH levels in male ERKOs appear to be regulated largely by nonsteroidal testicular factors such as inhibin. Finally, these data suggest that hypothalamic ERbeta may not be involved in mediating the negative feedback effects of T on serum LH and FSH in male mice.


Assuntos
Hormônio Foliculoestimulante/sangue , Hormônios Esteroides Gonadais/farmacologia , Hormônio Liberador de Gonadotropina/análise , Hipotálamo/química , Hormônio Luteinizante/sangue , Receptores de Estrogênio/fisiologia , Animais , Castração , Estradiol/sangue , Hormônio Foliculoestimulante/genética , Hormônio Luteinizante/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/análise
19.
Steroids ; 63(5-6): 352-4, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9618801

RESUMO

Several reports have shown an interaction between the estrogen receptor (ER) and the protein kinase C (PKC) intracellular pathways. Data from our laboratory showed that PKC activation can modulate ER levels and responsiveness in estrogen target tissues such as uterus and bone. In particular, ROS.SMER #14 osteoblastic cells, stably transfected with the mouse ER, undergo specific morphological changes in vitro. ROS.SMER #14 cells at post-confluence express a differentiated phenotype and become unresponsive to estrogenic stimulation. Interestingly, ER mRNA and protein levels were not modified by post-confluence, but ER binding sites/cell (2500-3000/cell at subconfluence) were undetectable. Moreover, PKC activity was significantly increased in post-confluent cells. Inhibition of PKC by H7 or staurosporin (PKC inhibitors) or down-regulation by long-term treatment with 12-O-tetradecanoylphorbol-13-acetate enchanced ER binding capacity in a dose-dependent manner. Since the PKC family includes several different isoforms that play different roles in cell homeostasis, we evaluated whether specific isoenzymes were involved in this event. To address this question, Western blotting analysis was performed on both sub- and post-confluent ROS.SMER #14 cells using antibodies against different PKC isoforms. In conclusion, our preliminary data indicate that estrogen responsiveness of osteoblastic cells can be highly regulated by PKC. Finally, these data suggest that this intracellular interaction might play an important role in modulating hormonal and pharmacological responsiveness of bone tissue.


Assuntos
Osteoblastos/fisiologia , Proteína Quinase C/fisiologia , Receptores de Estrogênio/fisiologia , Animais , Diferenciação Celular , Inibidores Enzimáticos/farmacologia , Camundongos , Osteoblastos/citologia , Proteína Quinase C/antagonistas & inibidores
20.
Proc Natl Acad Sci U S A ; 94(21): 11540-5, 1997 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-9326645

RESUMO

An essential component of regulated steroidogenesis is the translocation of cholesterol from the cytoplasm to the inner mitochondrial membrane where the cholesterol side-chain cleavage enzyme carries out the first committed step in steroidogenesis. Recent studies showed that a 30-kDa mitochondrial phosphoprotein, designated steroidogenic acute regulatory protein (StAR), is essential for this translocation. To allow us to explore the roles of StAR in a system amenable to experimental manipulation and to develop an animal model for the human disorder lipoid congenital adrenal hyperplasia (lipoid CAH), we used targeted gene disruption to produce StAR knockout mice. These StAR knockout mice were indistinguishable initially from wild-type littermates, except that males and females had female external genitalia. After birth, they failed to grow normally and died from adrenocortical insufficiency. Hormone assays confirmed severe defects in adrenal steroids-with loss of negative feedback regulation at hypothalamic-pituitary levels-whereas hormones constituting the gonadal axis did not differ significantly from levels in wild-type littermates. Histologically, the adrenal cortex of StAR knockout mice contained florid lipid deposits, with lesser deposits in the steroidogenic compartment of the testis and none in the ovary. The sex-specific differences in gonadal involvement support a two-stage model of the pathogenesis of StAR deficiency, with trophic hormone stimulation inducing progressive accumulation of lipids within the steroidogenic cells and ultimately causing their death. These StAR knockout mice provide a useful model system in which to determine the mechanisms of StAR's essential roles in adrenocortical and gonadal steroidogenesis.


Assuntos
Corticosteroides/farmacologia , Hiperplasia Suprarrenal Congênita/genética , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Glândulas Suprarrenais/patologia , Hiperplasia Suprarrenal Congênita/sangue , Hiperplasia Suprarrenal Congênita/patologia , Hormônio Adrenocorticotrópico/sangue , Aldosterona/sangue , Animais , Animais Recém-Nascidos , Corticosterona/sangue , Hormônio Liberador da Corticotropina/sangue , Dexametasona/análogos & derivados , Dexametasona/farmacologia , Modelos Animais de Doenças , Embrião de Mamíferos , Feminino , Fludrocortisona/farmacologia , Hormônio Foliculoestimulante/sangue , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Hipotálamo/metabolismo , Metabolismo dos Lipídeos , Lipídeos/análise , Masculino , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Ovário/patologia , Ratos , Caracteres Sexuais , Testículo/patologia , Testosterona/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...