Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Brain Struct Funct ; 224(8): 2983-2999, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31485718

RESUMO

Guanylyl cyclase C (GUCY2C) is the afferent central receptor in the gut-brain endocrine axis regulated by the anorexigenic intestinal hormone uroguanylin. GUCY2C mRNA and protein are produced in the hypothalamus, a major center regulating appetite and metabolic homeostasis. Further, GUCY2C mRNA and protein are expressed in the ventral midbrain, a principal structure regulating hedonic reward from behaviors including eating. While GUCY2C is expressed in hypothalamus and midbrain, its precise neuroanatomical organization and relationship with circuits regulating satiety remain unknown. Here, we reveal that hypothalamic GUCY2C mRNA is confined to the ventral premammillary nucleus (PMV), while in midbrain it is produced by neurons in the ventral tegmental area (VTA) and substantia nigra (SN). GUCY2C in the PMV is produced by 46% of neurons expressing anorexigenic leptin receptors, while in the VTA/SN it is produced in most tyrosine hydroxylase-immunoreactive neurons. In contrast to mRNA, GUCY2C protein is widely distributed throughout the brain in canonical sites of PMV and VTA/SN axonal projections. Selective stereotaxic ablation of PMV or VTA/SN neurons eliminated GUCY2C only in their respective canonical projection sites. Conversely, specific anterograde tracer analyses of PMV or VTA/SN neurons confirmed distinct GUCY2C-immunoreactive axons projecting to those canonical locations. Together, these findings reveal two discrete neuronal circuits expressing GUCY2C originating in the PMV in the hypothalamus and in the VTA/SN in midbrain, which separately project to other sites throughout the brain. They suggest a structural basis for a role for the GUCY2C-uroguanylin gut-brain endocrine axis in regulating homeostatic and behavioral components contributing to satiety.


Assuntos
Hipotálamo Posterior/metabolismo , Receptores de Enterotoxina/análise , Substância Negra/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Axônios , Feminino , Hipotálamo Posterior/citologia , Masculino , Camundongos Endogâmicos C57BL , Vias Neurais/citologia , RNA Mensageiro/análise , Substância Negra/citologia , Área Tegmentar Ventral/citologia
2.
Neuroscience ; 295: 23-38, 2015 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-25796140

RESUMO

Exercise reduces the risk of developing a number of neurological disorders and increases the efficiency of cellular energy production. However, overly strenuous exercise produces oxidative stress. Proper oxygenation is crucial for the health of all tissues, and tight regulation of cellular oxygen is critical to balance O2 levels and redox homeostasis in the brain. Hypoxia Inducible Factor (HIF)1α and HIF2α are transcription factors regulated by cellular oxygen concentration that initiate gene regulation of vascular development, redox homeostasis, and cell cycle control. HIF1α and HIF2α contribute to important adaptive mechanisms that occur when oxygen and ROS homeostasis become unbalanced. It has been shown that preconditioning by exposure to a stressor prior to a hypoxic event reduces damage that would otherwise occur. Previously we reported that 3 months of exercise protects SNpc dopaminergic (DA) neurons from toxicity caused by Complex I inhibition. Here, we identify the cells in the SNpc that express HIF1α and HIF2α and show that running exercise produces hypoxia in SNpc DA neurons, and alters the expression of HIF1α and HIF2α. In mice carrying a conditional knockout of Hif1α in postnatal neurons we observe that exercise alone produces SNpc TH+ DA neuron loss. Loss of HIF1α also abolishes exercise-induced neuroprotection. In mice lacking Hif2α in postnatal neurons, the number of TH+ DA neurons in the adult SNpc is diminished, but 3months of exercise rescues this loss. We conclude that HIF1α is necessary for exercise-induced neuroprotection and both HIF1α and HIF2α are necessary for the survival and function of adult SNpc DA neurons.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Neurônios Dopaminérgicos/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Intoxicação por MPTP/reabilitação , Parte Compacta da Substância Negra/citologia , Condicionamento Físico Animal/métodos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Sobrevivência Celular/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Intoxicação por MPTP/induzido quimicamente , Intoxicação por MPTP/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , RNA Mensageiro/metabolismo , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , beta-Galactosidase/metabolismo
3.
Neuroscience ; 214: 84-105, 2012 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-22542874

RESUMO

Parkinson's disease (PD) is a progressive neurodegenerative disorder whose etiology is thought to have environmental (toxin) and genetic contributions. The neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyrimidine (MPTP) induces pathological features of PD including loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and striatal dopamine (DA) depletion. We previously described the striatal transcriptional response following acute MPTP administration in MPTP-sensitive C57BL/6J mice. We identified three distinct phases: early (5h), intermediate (24h) and late (72h) and reported that the intermediate and late responses were absent in MPTP-resistant Swiss-Webster (SWR) mice. Here we show that C57BL/6J mice pre-treated with a single 40 mg/kg dose of MPTP and treated 9 days later with 4×20 mg/kg MPTP, display a striatal transcriptional response similar to that of MPTP-resistant SWR mice, i.e. a robust acute response but no intermediate or late response. Transcriptional refractoriness is dependent upon the dose of the priming challenge with as little as 10mg/kg MPTP being effective and can persist for more than 28 days. Priming of SWR mice has no effect on their response to subsequent challenge with MPTP. We also report that paraquat, another free radical producer, also elicits striatal transcriptional alterations but these are largely distinct from those triggered by MPTP. Paraquat-induced changes are also refractory to priming with paraquat. However neither paraquat nor MPTP elicits cross-attenuation. Thus exposure to specific toxins triggers distinct transcriptional responses in striatum that are influenced by prior exposure to the same toxin. The prolonged refractory period described here for MPTP could explain at the molecular level the reported discrepancies between different MPTP administration regimens and may have implications for our understanding of the relationship between environmental toxin exposure and PD.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/administração & dosagem , Transtornos Parkinsonianos/genética , Transcrição Gênica/fisiologia , Animais , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/fisiologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Transtornos Parkinsonianos/induzido quimicamente , Fatores de Tempo , Transcrição Gênica/efeitos dos fármacos
4.
Neuroscience ; 161(4): 1082-90, 2009 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-19376196

RESUMO

The substantia nigra pars compacta (SNpc) is a compact brain structure that contains a variable distribution of cells in both medial to lateral and rostral to caudal dimensions. The SNpc is the primary brain structure affected in Parkinson's disease, where loss of dopaminergic neurons is one of the major hallmarks of the disorder. Neurotoxic and genetic models of Parkinson's disease, as well as mechanisms to treat this disorder, are modeled in the mouse. To accurately assess the validity of a model, one needs to be assured that the method(s) of analysis is accurate. Here, we determined the total number of dopaminergic neurons in the SNpc of the C57BL/6J mouse by serial reconstruction then compared that value to estimates derived using model-based stereology and design-based stereology. Serial reconstruction of the SNpc revealed the total number of SNpc dopaminergic neurons to be 8305+/-540 (+/-SEM). We compared this empirically derived neuron number to model based and design-based stereological estimates. We found that model based estimates gave a value of 8002+/-91 (+/-SEM) while design-based estimates were 8716+/-338 (+/-SEM). Statistical analysis showed no significant difference between estimates generated using model- or design-based stereological methods compared to empirically-derived counts using serial reconstruction.


Assuntos
Dopamina/metabolismo , Imageamento Tridimensional/métodos , Modelos Neurológicos , Neurônios/citologia , Neurônios/metabolismo , Substância Negra/citologia , Análise de Variância , Animais , Contagem de Células , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal
5.
Neuroscience ; 145(2): 654-68, 2007 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-17258864

RESUMO

Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). With the exception of a few rare familial forms of the disease, the precise molecular mechanisms underlying PD are unknown. Inflammation is a common finding in the PD brain, but due to the limitation of postmortem analysis its relationship to disease progression cannot be established. However, studies using the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of PD have also identified inflammatory responses in the nigrostriatal pathway that precede neuronal degeneration in the SNpc. To assess the pathological relevance of these inflammatory responses and to identify candidate genes that might contribute to neuronal vulnerability, we used quantitative reverse-transcription polymerase chain reaction (qRT-PCR) to measure mRNA levels of 11 cytokine and chemokine encoding genes in the striatum of MPTP-sensitive (C57BL/6J) and MPTP-insensitive (Swiss Webster, SWR) mice following administration of MPTP. The mRNA levels of all 11 genes changed following MPTP treatment, indicating the presence of inflammatory responses in both strains. Furthermore, of the 11 genes examined only 3, interleukin 6 (Il-6), macrophage inflammatory protein 1 alpha/CC chemokine ligand 3 (Mip-1alpha/Ccl3) and macrophage inflammatory protein 1 beta/CC chemokine ligand 4 (Mip-1beta/Ccl4), were differentially regulated between C57BL/6J and SWR mice. In both mouse strains, the level of monocyte chemoattractant protein 1/CC chemokine ligand 2 (Mcp-1/Ccl2) mRNA was the first to increase following MPTP administration, and might represent a key initiating component of the inflammatory response. Using Mcp-1/Ccl2 knockout mice backcrossed onto a C57BL/6J background we found that MPTP-stimulated Mip-1alpha/Ccl3 and Mip-1beta/Ccl4 mRNA expression was significantly lower in the knockout mice; suggesting that Mcp-1/Ccl2 contributes to MPTP-enhanced expression of Mip-1alpha/Ccl3 and Mip-1beta/Ccl4. However, stereological analysis of SNpc neuronal loss in Mcp-1/Ccl2 knockout and wild-type mice showed no differences. These findings suggest that it is the ability of dopaminergic SNpc neurons to survive an inflammatory insult, rather than genetically determined differences in the inflammatory response itself, that underlie the molecular basis of MPTP resistance.


Assuntos
Encéfalo/metabolismo , Encefalite/genética , Mediadores da Inflamação/metabolismo , Proteínas do Tecido Nervoso/genética , Transtornos Parkinsonianos/genética , RNA Mensageiro/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Animais , Encéfalo/fisiopatologia , Sobrevivência Celular/genética , Quimiocinas/genética , Corpo Estriado/metabolismo , Corpo Estriado/fisiopatologia , Citocinas/genética , Modelos Animais de Doenças , Encefalite/metabolismo , Encefalite/fisiopatologia , Feminino , Regulação da Expressão Gênica/fisiologia , Predisposição Genética para Doença/genética , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Degeneração Neural/genética , Degeneração Neural/metabolismo , Degeneração Neural/fisiopatologia , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/fisiopatologia , Especificidade da Espécie , Fatores de Tempo
6.
Neuroscience ; 137(3): 905-13, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16298078

RESUMO

Cocaine abuse is a significant problem in the United States, including its use by approximately 1% of pregnant women. Cocaine acts as an indirect agonist of dopamine at the dopamine transporter, resulting in the presence of excess dopamine in the synapse. Since synaptic dopamine can rapidly oxidize to form free radicals, it was hypothesized that exposure to this drug might produce damage in dopaminergic systems such as the substantia nigra pars compacta, damage to which is a hallmark of Parkinson's disease. To test this hypothesis we exposed mice both in utero and as adults to cocaine and examined its effects on the nigrostriatal system. We found that exposure to cocaine both in utero or as adults did not affect substantia nigra cell number, but did make these neurons more susceptible to the parkinsonian toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. We also found long-lasting changes in D2 receptor mRNA levels as well as changes in the monoamine transport system and several growth factors. This work suggests that use of cocaine might be a predisposing factor for development of Parkinson's disease in both adults exposed chronically as well as in individuals exposed prenatally.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Cocaína/farmacologia , Dopaminérgicos/toxicidade , Inibidores da Captação de Dopamina/farmacologia , Doença de Parkinson Secundária/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/antagonistas & inibidores , Animais , Contagem de Células , Primers do DNA , Feminino , Imuno-Histoquímica , Camundongos , Doença de Parkinson Secundária/genética , Doença de Parkinson Secundária/patologia , Gravidez , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de Dopamina D2/biossíntese , Receptores de Dopamina D2/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Neurotox Res ; 7(3): 193-202, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15897154

RESUMO

Parkinson disease (PD) is a common neurodegenerative disease of unknown origin that is characterized, mainly, by a significant reduction in the number of dopamine neurons in the substantia nigra pars compacta (SNpc) of the brain and a dramatic reduction in dopamine levels in the corpus striatum. For reasons that we do not know, the dopamine neuron seems to be more vulnerable to damage than any other neuron in the brain. Although hypotheses of damage to the dopamine neuron include oxidative stress, growth factor decline, excitotoxicity, inflammation in the SNpc and protein aggregation, oxidative stress in the nigrostriatal dopaminergic system garners a significant amount of attention. In the oxidative stress hypothesis of PD, superoxide, nitric oxide and dopamine all conspire to create an environment that can be detrimental to the dopamine neuron. MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine), the tool of choice for investigations into the mechanisms involved in the death of dopamine neurons in PD, has been used extensively in attempts to sort out what happens in and around the dopamine neuron. Herein, we review the roles of dopamine, superoxide and nitric oxide in the demise of the dopamine neuron in the MPTP model of PD as it relates to the death of the dopamine neuron noted in PD.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/metabolismo , Dopamina/metabolismo , Neurônios/metabolismo , Óxido Nítrico/metabolismo , Substância Negra/citologia , Superóxidos/metabolismo , Animais , Meio Ambiente , Humanos , Redes Neurais de Computação , Neuroglia/efeitos dos fármacos , Neuroglia/fisiologia , Neurônios/efeitos dos fármacos , Substância Negra/metabolismo
8.
Nat Genet ; 29(4): 404-11, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11726927

RESUMO

Somatic inactivation of PTEN occurs in different human tumors including glioblastoma, endometrial carcinoma and prostate carcinoma. Germline mutations in PTEN result in a range of phenotypic abnormalities that occur with variable penetrance, including neurological features such as macrocephaly, seizures, ataxia and Lhermitte-Duclos disease (also described as dysplastic gangliocytoma of the cerebellum). Homozygous deletion of Pten causes embryonic lethality in mice. To investigate function in the brain, we used Cre-loxP technology to selectively inactivate Pten in specific mouse neuronal populations. Loss of Pten resulted in progressive macrocephaly and seizures. Neurons lacking Pten expressed high levels of phosphorylated Akt and showed a progressive increase in soma size without evidence of abnormal proliferation. Cerebellar abnormalities closely resembled the histopathology of human Lhermitte-Duclos disease. These results indicate that Pten regulates neuronal size in vivo in a cell-autonomous manner and provide new insights into the etiology of Lhermitte-Duclos disease.


Assuntos
Tamanho Celular/genética , Doenças Cerebelares/genética , Genes Supressores de Tumor , Neurônios/patologia , Monoéster Fosfórico Hidrolases/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Ciclo Celular/genética , Divisão Celular/genética , Doenças Cerebelares/patologia , Inibidor de Quinase Dependente de Ciclina p27 , Modelos Animais de Doenças , Deleção de Genes , Proteína Glial Fibrilar Ácida/genética , Imuno-Histoquímica , Integrases/genética , Camundongos , Camundongos Transgênicos , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/genética , Fosforilação , Convulsões/genética , Proteínas Supressoras de Tumor/genética , Proteínas Virais/genética
9.
Glia ; 34(2): 73-80, 2001 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11307156

RESUMO

Parkinson's disease (PD) is a debilitating neurological disorder that strikes approximately 2% of people over age 50. Current hypotheses propose that the cause of PD is multifactorial, involving environmental agents and genetic predisposition. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induces parkinsonism in many species, including humans and shows strain specificity in mice. The mechanism of strain specificity, however, remains unknown. Using novel chimeric murine substantia nigra cultures, we demonstrate that sensitivity to MPTP is conferred by glia and that it does not involve the MAO-B conversion of MPTP to MPP(+). C57Bl/6J dopaminergic neurons exposed to MPP(+) demonstrated a 39% loss when cultured on C57Bl/6J glia compared with 17% neuron loss when cultured on resistant SWR/J glia. Similarly, SWR/J neurons exposed to MPP(+) demonstrated a 4% loss when cultured on SWR/J glia, but a 14% loss when cultured on sensitive C57Bl/6J glia. The identification of glia as the critical cell type in the genesis of experimental Parkinsonism provides a target for the development of new anti-parkinsonian therapies.


Assuntos
Neuroglia/efeitos dos fármacos , Transtornos Parkinsonianos/patologia , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , 1-Metil-4-fenilpiridínio/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Células Cultivadas , Dopamina/farmacologia , Dopaminérgicos/farmacologia , Herbicidas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/citologia , Neuroglia/metabolismo , Transtornos Parkinsonianos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Especificidade da Espécie , Substância Negra/citologia
10.
J Recept Signal Transduct Res ; 20(1): 87-103, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10711499

RESUMO

In adult animals, signaling through the leptin receptor (OB-R) has been shown to play a critical role in fat metabolism. However, it is not known when these receptors are first expressed and what their role may be during embryonic development. To date, at least 6 splice variants of the OB-R have been identified. Although the function of each of these individual splice variants are unknown, only one of them, ob-rL ,encodes a receptor with a long intracellular domain that is implicated in OB-R signaling. In this study we have used in situ hybridization to examine the localization of OB-R splice variants during embryonic development of C57B1/6J mice. Using a probe, ob-r, that recognizes all of the splice variants, ob-r mRNA was found to be distributed in developing bone, mesenchyme, notochord and liver. In addition, epithelial structures including leptomeninges, choroid plexi and hair follicles also expressed ob-r. No ob-r mRNA was detected in the CNS. ob-rL, expression was only detected in notochord, bone and mesenchyme. The differential expression of these two mRNA isoforms suggests that the extracellular and intracellular domains of the OB receptor perform different biological functions.


Assuntos
Proteínas de Transporte/genética , Feto/metabolismo , RNA Mensageiro/análise , Receptores de Superfície Celular , Animais , Feminino , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Splicing de RNA , Receptores para Leptina
11.
Proc Natl Acad Sci U S A ; 96(23): 13462-7, 1999 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-10557343

RESUMO

Development of the central nervous system requires proliferation of neuronal and glial cell precursors followed by their subsequent differentiation in a highly coordinated manner. The timing of neuronal cell cycle exit and differentiation is likely to be regulated in part by inhibitors of cyclin-dependent kinases. Overlapping and sustained patterns of expression of two cyclin-dependent kinases, p19(Ink4d) and p27(Kip1), in postmitotic brain cells suggested that these proteins may be important in actively repressing neuronal proliferation. Animals derived from crosses of Ink4d- null with Kip1-null mice exhibited bradykinesia, proprioceptive abnormalities, and seizures, and died at about 18 days after birth. Metabolic labeling of live animals with bromodeoxyuridine at postnatal days 14 and 18, combined with immunolabeling of neuronal markers, showed that subpopulations of central nervous system neurons were proliferating in all parts of the brain, including normally dormant cells of the hippocampus, cortex, hypothalamus, pons, and brainstem. These cells also expressed phosphorylated histone H3, a marker for late G(2) and M-phase progression, indicating that neurons were dividing after they had migrated to their final positions in the brain. Increased proliferation was balanced by cell death, resulting in no gross changes in the cytoarchitecture of the brains of these mice. Therefore, p19(Ink4d) and p27(Kip1) cooperate to maintain differentiated neurons in a quiescent state that is potentially reversible.


Assuntos
Proteínas de Transporte/genética , Proteínas de Ciclo Celular , Divisão Celular/genética , Inibidor p16 de Quinase Dependente de Ciclina , Quinases Ciclina-Dependentes/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/genética , Neurônios/citologia , Proteínas Supressoras de Tumor , Animais , Apoptose/genética , Encéfalo/citologia , Diferenciação Celular , Inibidor de Quinase Dependente de Ciclina p19 , Inibidor de Quinase Dependente de Ciclina p27 , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
12.
Brain Res Mol Brain Res ; 70(1): 159-63, 1999 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-10381555

RESUMO

In this study, we examined the levels of activated caspase-3 in the kainic acid (KA) model of hippocampal degeneration in both sensitive (FVB/N) and resistant (129/SvEMS) strains of mice. At 30 h, 2 and 4 days following KA administration, animals were sacrificed and brains examined for pyknosis, TUNEL labeling, and activated caspase-3 immunoreactivity. Catalytically active caspase-3 was first detected 30 h following KA treatment in the sensitive, FVB/N strain. This was 18 h before the appearance of pyknosis or TUNEL labeling. The expression of activated caspase-3 continues up to 4 days post-injection. No activated caspase-3 immunoreactivity was detected in the resistant, 129/SvEMS strain, neither was there evidence of pyknosis or TUNEL staining. This suggests that activation of caspase-3 is a necessary component of KA-induced cell death.


Assuntos
Apoptose/efeitos dos fármacos , Caspases/fisiologia , Agonistas de Aminoácidos Excitatórios/toxicidade , Hipocampo/efeitos dos fármacos , Ácido Caínico/toxicidade , Proteínas do Tecido Nervoso/fisiologia , Neurônios/patologia , Animais , Caspase 3 , Resistência a Medicamentos , Ativação Enzimática/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/patologia , Marcação In Situ das Extremidades Cortadas , Ácido Caínico/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos , Convulsões/induzido quimicamente , Convulsões/enzimologia , Convulsões/patologia
13.
Brain Res ; 828(1-2): 91-103, 1999 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10320728

RESUMO

1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) has been used as a potent neurotoxin to approximate, in animals, the pathology that is observed in human Parkinson's disease. In this study, we examine the toxicity of MPTP in seven strains of mice, spanning a genetic continuum of Mus musculus as a prelude to uncovering complex traits associated with MPTP toxicity. Seven days following injection of 80 mg/kg MPTP (4x20 mg/kg every 2 h), we find that the individual mouse strains exhibit dramatic differences in SNpc neuron survival, ranging from 63% cell loss in C57BL/6J mice to 14% cell loss in Swiss-Webster (SW) mice. In order to determine if the susceptibility trait was dominant, additive or recessive, we crossed C57Bl/6J mice with either SWR/J or AKR/J mice and examined the effect of MPTP on F1 C57BL/6JxSWR/J or F1 C57BL/6JxAKR/J animals. We find that all of the F1 animals were phenotypically identical to the C57BL/6J animals. In addition, no gender differences were noted in any of the MPTP-treated inbred mice or in the F1 animals. These results suggest that susceptibility to cell loss following MPTP is autosomal dominant and this polymorphism is carried on the C57BL/6J allele.


Assuntos
Dopaminérgicos/toxicidade , Genes Dominantes , Intoxicação por MPTP , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/genética , Animais , Gânglios da Base/citologia , Gânglios da Base/efeitos dos fármacos , Gânglios da Base/fisiopatologia , Contagem de Células , Suscetibilidade a Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Fatores Sexuais , Especificidade da Espécie , Substância Negra/citologia , Substância Negra/efeitos dos fármacos , Substância Negra/fisiopatologia , Tirosina 3-Mono-Oxigenase/análise
14.
J Recept Signal Transduct Res ; 19(1-4): 245-66, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10071762

RESUMO

A high affinity receptor for OB protein was recently cloned from the choroid plexus of mice. At least six alternatively spliced forms of the OB receptor (OB-R) gene have been described, all of which encode proteins containing the OB-R extracellular domain. One splice variant encodes a receptor with a long intracellular domain, OB-RL, that has been implicated in OB-R signaling. Here, we have used in situ hybridization to examine the localization of OB-R splice variants in brain and peripheral tissues of adult and newborn mice. Using a probe hybridizing with all known splice variants, we confirmed that OB-R mRNA was widely distributed in the adult tissues. In the CNS, choroid plexus was the major site of expression. We now demonstrate that OB-R mRNA is expressed in peripheral tissues; primarily associated with connective tissues. In addition, OB-R mRNA was detected at higher levels in peripheral tissues of newborn mice than in adult mice. With a probe specific for OB-RL, we confirmed that high mRNA expression was detected in hypothalamic nuclei, while low levels were observed in choroid plexus. We now report that in peripheral tissues of adult mice, OB-RL mRNA expression was either very low or undetectable. In newborn mice, the pattern of OB-RL message expression in the CNS was similar to that of adult mice, while bone was the site of highest OB-RL message expression in the peripheral tissue. These data suggest different biological roles for OB-R splice variants encoding the short and long forms of OB-R. The localization of OB-RL to hypothalamic nuclei supports the idea that OB-RL is the brain receptor that mediates OB protein signaling and actions. In addition, the expression of OB-R message in newborn mice also suggests a biological role of OB-R during development in mice.


Assuntos
Encéfalo/metabolismo , Proteínas de Transporte/genética , Variação Genética , Receptores de Superfície Celular , Processamento Alternativo , Animais , Animais Recém-Nascidos , Sequência de Bases , Primers do DNA/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores para Leptina , Distribuição Tecidual
15.
Hum Mol Genet ; 7(11): 1787-94, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9736781

RESUMO

Protective protein/cathepsin A (PPCA) is a pleiotropic lysosomal enzyme that complexes with beta-galactosidase and neuraminidase, and possesses serine carboxypeptidase activity. Its deficiency in man results in the neurodegenerative lysosomal storage disorder galactosialidosis (GS). The mouse model of this disease resembles the human early onset phenotype and results in severe nephropathy and ataxia. To understand better the pathophysiology of the disease, we compared the occurrence of lysosomal PPCA mRNA and protein in normal adult mouse tissues with the incidence of lysosomal storage in PPCA(-/-) mice. PPCA expression was markedly variable among different tissues. Most sites that produced both mRNA and protein at high levels in normal mice showed extensive and overt storage in the knockout mice. However, this correlation was not consistent as some cells that normally expressed high levels of PPCA were unaffected in their storage capability in the PPCA(-/-) mice. In addition, some normally low expressing cells accumulated large amounts of undegraded products in the GS mouse. This apparent discrepancy may reflect a requirement for the catalytic rather than the protective function of PPCA and/or the presence of cell-specific substrates in certain cell types. A detailed map showing the cellular distribution of PPCA in nomal mouse tissues as well as the sites of lysosomal storage in deficient mice is critical for accurate assessment of the effects of therapeutic interventions.


Assuntos
Carboxipeptidases/genética , Carboxipeptidases/metabolismo , Doenças por Armazenamento dos Lisossomos/genética , Doenças por Armazenamento dos Lisossomos/patologia , Animais , Encéfalo/enzimologia , Encéfalo/patologia , Catepsina A , Epididimo/enzimologia , Epididimo/patologia , Feminino , Imuno-Histoquímica , Hibridização In Situ , Intestinos/enzimologia , Intestinos/patologia , Fígado/enzimologia , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Ovário/enzimologia , Ovário/patologia , Baço/enzimologia , Baço/patologia , Testículo/enzimologia , Testículo/patologia , Útero/enzimologia , Útero/patologia
16.
Dev Biol ; 198(1): 57-81, 1998 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-9640332

RESUMO

The impact of the nerve growth factor (NGF) family of neurotrophins and their receptors was examined on the cutaneous innervation in the mystacial pads of mice. Ten sets of unmyelinated and thinly myelinated sensory and autonomic innervation were evaluated that terminated in the epidermis, upper dermis, and upper part of the intervibrissal hair follicles. Mystacial pads were analyzed from newborn to 4-week-old mice that had homozygous functional deletions of the genes for NGF, brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), neurotrophin-4 (NT-4), tyrosine kinase (trk) A, trkB, trkC, or p75. Mystacial pads were also analyzed in adult transgenic mice that had overproduction of NGF, BDNF, or NT-3 driven by a keratin promoter gene. The innervation was revealed by using immunofluorescence and immunocytochemistry with antibodies for protein gene product (PGP) 9.5, calcitonin gene-related product (CGRP), substance P (SP), galanin (GAL), neuropeptide Y (NPY), tyrosine hydroxylase (TH), and a neurofilament protein. The cumulative results indicated that NGF/trkA signaling plays a major role in the outgrowth and proliferation of sensory axons, whereas NT-3/ trkA signaling plays a major role in the formation of sensory endings. TrkC is also essential for the development of three sets of trkA-dependent sensory innervation that coexpress CGRP, SP, and GAL. Another set of sensory innervation that only coexpressed CGRP and SP was solely dependent upon NGF and trkA. Surprisingly, most sets of trkA-dependent sensory innervation are suppressed by trkB perhaps interacting with p75. BDNF and NT-4 appear to mediate this suppressing effect in the upper dermis and NT-4 in the epidermis. In contrast to sensory innervation, sympathetic innervation to the necks of intervibrissal hair follicles depends upon NGF/trkA signaling interacting with p75 for both the axon outgrowth and ending formation. Although NT-3/trkA signaling is essential for the full complement of sympathetic neurons, NT-3 is detrimental to the formation of sympathetic terminations to the necks of hair follicles. TrkB signaling mediated by BDNF but not NT-4 also suppresses these sympathetic terminations. One sparse set of innervation, perhaps parasympathetic, terminating at the necks of hair follicles is dependent solely upon NT-3 and trkC. Taken together, our results indicate that the innervation of the epidermis, upper dermis, and the upper portion of hair follicles is regulated by a competitive balance between promoting and suppressing effects of the various neurotrophins.


Assuntos
Fatores de Crescimento Neural/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Pele/inervação , Animais , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento/genética , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Fatores de Crescimento Neural/genética , Neurotrofina 3 , Receptor de Fator de Crescimento Neural , Receptor trkA/genética , Receptor trkA/fisiologia , Receptores de Fator de Crescimento Neural , Transdução de Sinais/fisiologia , Pele/citologia
17.
Blood ; 90(5): 1777-86, 1997 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9292510

RESUMO

We have investigated the utility of the green fluorescent protein (GFP) to serve as a marker to assess retroviral gene transfer into hematopoietic cells and as a tool to identify and enrich for cells expressing high levels of the vector-encoded transcript. GFP, by virtue of a naturally occurring chromophore encoded in its primary sequence, displays autonomous fluorescence, thus eliminating the need for antibody or cytochemical staining to detect its expression. A bicistronic murine stem cell virus (MSCV)-based retroviral vector was constructed containing the GFP cDNA and a mutant, human dihydrofolate reductase gene. High-titer, ecotropic retroviral producer cells free of replication competent virus were generated and used to transduce murine bone marrow cells by cocultivation. Within 24 hours after completion of the transduction procedure, a high proportion (40% to 70%) of the marrow cells were intensely fluorescent compared to mock-transduced cells or cells transduced with a control retrovirus. Erythroid and myeloid hematopoietic colonies derived from GFP-transduced marrow were easily scored for retroviral gene transfer by direct in situ fluorescence microscopy. Clonogenic progenitors expressing increased levels of antifolate drug resistance could be enriched from the GFP-transduced marrow population by fluorescence activated cell sorting of cells expressing high levels of GFP. In vivo, splenic hematopoietic colonies and peripheral blood cells from animals transplanted with GFP-transduced marrow displayed intense fluorescence. These results show that GFP is an excellent marker for scoring and tracking gene-modified hematopoietic cells and for allowing rapid selection and enrichment of transduced cells expressing high levels of the transgene.


Assuntos
Células da Medula Óssea , Técnicas de Transferência de Genes , Hematopoese/genética , Proteínas Luminescentes/genética , Animais , Expressão Gênica , Marcadores Genéticos , Vetores Genéticos , Proteínas de Fluorescência Verde , Humanos , Camundongos , Retroviridae
18.
Brain Res Mol Brain Res ; 46(1-2): 333-7, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9191112

RESUMO

L7/pcp-2 is expressed exclusively in cerebellar Purkinje and retinal bipolar neurons. While the function of L7/pcp-2 is unknown, its mRNA is trafficked into dendrites, suggesting a role in dendritic physiology. To elucidate its function, L7/pcp-2-null mice were generated. These mice are neurologically normal with no signs of cerebellar or retinal dysfunction. The mice are indistinguishable from wild-type littermates with regards to gross neuroanatomy and fine structure of Purkinje cell dendrites.


Assuntos
Neuropeptídeos/fisiologia , Células de Purkinje/fisiologia , Retina/fisiologia , Animais , Fatores de Troca do Nucleotídeo Guanina , Camundongos , Camundongos Endogâmicos C57BL
19.
Perspect Dev Neurobiol ; 5(1): 33-41, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9509516

RESUMO

The advent of transgenic mouse technology and the harnessing of homologous recombination to eliminate genes in living animals have provided powerful new approaches to investigate some of the fundamental issues in developmental neurobiology. In this chapter we illustrate several examples of the use of these technologies to investigate the murine cerebellum. These range from using inert, marker transgenes to follow the organization strategy of the cerebellum, to eliminating specific cell populations and modifying the function and fate of Purkinje cells. Finally, we provide a perspective on future extensions of these transgenic approaches.


Assuntos
Cerebelo/embriologia , Animais , Mapeamento Encefálico , Comunicação Celular/fisiologia , Desenvolvimento Embrionário e Fetal/fisiologia , Marcadores Genéticos , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Células de Purkinje/fisiologia
20.
Brain Res ; 778(2): 251-64, 1997 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-9459542

RESUMO

G-protein-gated inward rectifying potassium channels (GIRKs) are a newly identified gene family. These gene products are thought to form functional channels through the assembly of heteromeric subunits. Recently, it has been demonstrated that a point mutation in the GIRK2 gene, one of the GIRK family members, is the cause of the neurological and reproductive defects observed in the weaver (wv) mutant mouse. The mechanism(s) by which a single amino acid substitution in GIRK2 protein leads to the severe phenotypes in the wv / wv mouse is not fully understood. However, it implicates the importance of GIRK channels in neuronal development. To characterize the mRNA expression patterns of GIRK1-3 during mouse brain development we have used in situ hybridization analyses. We found that the expression of all three genes showed developmental regulation. In most areas that showed expression, the levels of GIRK1-3 transcripts reached their peak at around postnatal day 10 (P10). In general, GIRK1 showed the least fluctuation in its levels of expression during development, while dynamic changes were found with the levels of GIRK2 and GIRK3 transcripts. GIRK3 becomes the predominant inward rectifying K+-channel in the brain at later postnatal ages. In the CNS regions affected in the wv / wv mouse, GIRK2 is the predominant inward rectifying channel that is expressed. This suggests that the presence of the other subtypes are able to compensate for the mutated GIRK2 channel in weaver neurons that survive.


Assuntos
Química Encefálica/genética , Camundongos Mutantes Neurológicos/anormalidades , Canais de Potássio Corretores do Fluxo de Internalização , Canais de Potássio/genética , Animais , Animais Recém-Nascidos , Cerebelo/química , Córtex Cerebral/química , Modelos Animais de Doenças , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Regulação da Expressão Gênica no Desenvolvimento , Hipocampo/química , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Doenças Neurodegenerativas/genética , Bulbo Olfatório/química , Gravidez , RNA Mensageiro/análise , Receptores Muscarínicos/genética , Substância Negra/química , Tálamo/química , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA