Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurochem ; 156(4): 481-498, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32583440

RESUMO

Glial cell line-derived neurotrophic factor (GDNF) has been reported to enhance dopaminergic neuron survival and differentiation in vitro and in vivo, although those results are still being debated. Glial cell line-derived neurotrophic factor (gdnf) is highly conserved in zebrafish and plays a role in enteric nervous system function. However, little is known about gdnf function in the teleost brain. Here, we employed clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 to impede gdnf function in the maintenance of dopaminergic neuron development. Genotyping of gdnf crispants revealed successful deletions of the coding region with various mutant band sizes and down-regulation of gdnf transcripts at 1, 3 and 7 day(s) post fertilization. Notably, ~20% reduction in ventral diencephalic dopaminergic neuron numbers in clusters 8 and 13 was observed in the gdnf-deficient crispants. In addition, gdnf depletion caused a modest reduction in dopaminergic neurogenesis as determined by 5-ethynyl-2'-deoxyuridine pulse chase assay. These deleterious effects could be partly attributed to deregulation of dopaminergic neuron fate specification-related transcription factors (otp,lmx1b,shha,and ngn1) in both crispants and established homozygous mutants with whole mount in-situ hybridization (WISH) on gdnf mutants showing reduced otpb and lmx1b.1 expression in the ventral diencephalon. Interestingly, locomotor function of crispants was only impacted at 7 dpf, but not earlier. Lastly, as expected, gdnf deficiency heightened crispants vulnerability to 1-methyl-4-phenylpyridinium toxic insult. Our results suggest conservation of teleost gdnf brain function with mammals and revealed the interactions between gdnf and transcription factors in dopaminergic neuron differentiation.


Assuntos
Diferenciação Celular/fisiologia , Diencéfalo/embriologia , Diencéfalo/metabolismo , Neurônios Dopaminérgicos/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Fatores de Transcrição/deficiência , Proteínas de Peixe-Zebra/deficiência , Animais , Animais Geneticamente Modificados , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fatores de Transcrição/genética , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
2.
J Neurosci Res ; 98(9): 1764-1779, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-31663646

RESUMO

Glial-derived neurotrophic factor (GDNF) has been proposed as a potent neurotrophic factor with the potential to cure neurodegenerative diseases. In the cochlea, GDNF has been detected in auditory neurons and sensory receptor cells and its expression is upregulated upon trauma. Moreover, the application of GDNF in different animal models of deafness has shown its capacity to prevent hearing loss and promoted its future use in therapeutic trials in humans. In the present study we have examined the endogenous requirement of GDNF during auditory development in mice. Using a lacZ knockin allele we have confirmed the expression of GDNF in the cochlea including its sensory regions during development. Global inactivation of GDNF throughout the hearing system using a Foxg1-Cre line causes perinatal lethality but reveals no apparent defects during formation of the cochlea. Using TrkC-Cre and Atoh1-Cre lines, we were able to generate viable mutants lacking GDNF in auditory neurons or both auditory neurons and sensory hair cells. These mutants show normal frequency-dependent auditory thresholds. However, mechanoelectrical response properties of outer hair cells (OHCs) in TrkC-Cre GDNF mutants are altered at low thresholds. Furthermore, auditory brainstem wave analysis shows an abnormal increase of wave I. On the other hand, Atoh1-Cre GDNF mutants show normal OHC function but their auditory brainstem wave pattern is reduced at the levels of wave I, III and IV. These results show that GDNF expression during the development is required to maintain functional hearing at different levels of the auditory system.


Assuntos
Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Audição/fisiologia , Animais , Limiar Auditivo , Cóclea/metabolismo , Orelha Interna/metabolismo , Potenciais Evocados Auditivos do Tronco Encefálico , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Células Ciliadas Auditivas/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
3.
Am J Physiol Renal Physiol ; 313(3): F576-F584, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28490528

RESUMO

Recently, new methods for assessing renal function in conscious mice (transcutaneous assessment) and for counting and sizing all glomeruli in whole kidneys (MRI) have been described. In the present study, these methods were used to assess renal structure and function in aging mice, and in mice born with a congenital low-nephron endowment. Age-related nephron loss was analyzed in adult C57BL/6 mice (10-50 wk of age), and congenital nephron deficit was assessed in glial cell line-derived neurotrophic factor heterozygous (GDNF HET)-null mutant mice. Renal function was measured through the transcutaneous quantitation of fluorescein isothiocyanate-sinistrin half-life (t1/2) in conscious mice. MRI was used to image, count, and size cationic-ferritin labeled glomeruli in whole kidneys ex vivo. Design-based stereology was used to validate the MRI measurements of glomerular number and mean volume. In adult C57BL/6 mice, older age was associated with fewer and larger glomeruli, and a rightward shift in the glomerular size distribution. These changes coincided with a decrease in renal function. GNDF HET mice had a congenital nephron deficit that was associated with glomerular hypertrophy and exacerbated by aging. These findings suggest that glomerular hypertrophy and hyperfiltration are compensatory processes that can occur in conjunction with both age-related nephron loss and congenital nephron deficiency. The combination of measurement of renal function in conscious animals and quantitation of glomerular number, volume, and volume distribution provides a powerful new tool for investigating aspects of renal aging and functional changes.


Assuntos
Envelhecimento/patologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Nefropatias/patologia , Nefropatias/fisiopatologia , Testes de Função Renal , Glomérulos Renais/patologia , Imageamento por Ressonância Magnética , Néfrons/anormalidades , Fatores Etários , Animais , Modelos Animais de Doenças , Fluoresceínas/administração & dosagem , Fluoresceínas/farmacocinética , Corantes Fluorescentes/administração & dosagem , Corantes Fluorescentes/farmacocinética , Predisposição Genética para Doença , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Taxa de Filtração Glomerular , Meia-Vida , Heterozigoto , Hipertrofia , Nefropatias/congênito , Glomérulos Renais/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligossacarídeos/administração & dosagem , Oligossacarídeos/farmacocinética , Fenótipo , Valor Preditivo dos Testes
4.
J Neurosci ; 37(6): 1581-1590, 2017 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-28096470

RESUMO

Midbrain dopamine neuron dysfunction contributes to various psychiatric and neurological diseases, including drug addiction and Parkinson's disease. Because of its well established dopaminotrophic effects, the therapeutic potential of glial cell line-derived neurotrophic factor (GDNF) has been studied extensively in various disorders with disturbed dopamine homeostasis. However, the outcomes from preclinical and clinical studies vary, highlighting a need for a better understanding of the physiological role of GDNF on striatal dopaminergic function. Nevertheless, the current lack of appropriate animal models has limited this understanding. Therefore, we have generated novel mouse models to study conditional Gdnf deletion in the CNS during embryonic development and reduction of striatal GDNF levels in adult mice via AAV-Cre delivery. We found that both of these mice have reduced amphetamine-induced locomotor response and striatal dopamine efflux. Embryonic GDNF deletion in the CNS did not affect striatal dopamine levels or dopamine release, but dopamine reuptake was increased due to increased levels of both total and synaptic membrane-associated dopamine transporters. Collectively, these results suggest that endogenous GDNF plays an important role in regulating the function of dopamine transporters in the striatum.SIGNIFICANCE STATEMENT Delivery of ectopic glial cell line-derived neurotrophic factor (GDNF) promotes the function, plasticity, and survival of midbrain dopaminergic neurons, the dysfunction of which contributes to various neurological and psychiatric diseases. However, how the deletion or reduction of GDNF in the CNS affects the function of dopaminergic neurons has remained unknown. Using conditional Gdnf knock-out mice, we found that endogenous GDNF affects striatal dopamine homeostasis and regulates amphetamine-induced behaviors by regulating the level and function of dopamine transporters. These data regarding the physiological role of GDNF are relevant in the context of neurological and neurodegenerative diseases that involve changes in dopamine transporter function.


Assuntos
Anfetamina/farmacologia , Encéfalo/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Inibidores da Captação de Dopamina/farmacologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Animais , Encéfalo/efeitos dos fármacos , Proteínas da Membrana Plasmática de Transporte de Dopamina/antagonistas & inibidores , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Distribuição Aleatória
5.
J Clin Invest ; 125(5): 1873-85, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25822020

RESUMO

Parkin and the glial cell line-derived neurotrophic factor (GDNF) receptor RET have both been independently linked to the dopaminergic neuron degeneration that underlies Parkinson's disease (PD). In the present study, we demonstrate that there is genetic crosstalk between parkin and the receptor tyrosine kinase RET in two different mouse models of PD. Mice lacking both parkin and RET exhibited accelerated dopaminergic cell and axonal loss compared with parkin-deficient animals, which showed none, and RET-deficient mice, in which we found moderate degeneration. Transgenic expression of parkin protected the dopaminergic systems of aged RET-deficient mice. Downregulation of either parkin or RET in neuronal cells impaired mitochondrial function and morphology. Parkin expression restored mitochondrial function in GDNF/RET-deficient cells, while GDNF stimulation rescued mitochondrial defects in parkin-deficient cells. In both cases, improved mitochondrial function was the result of activation of the prosurvival NF-κB pathway, which was mediated by RET through the phosphoinositide-3-kinase (PI3K) pathway. Taken together, these observations indicate that parkin and the RET signaling cascade converge to control mitochondrial integrity and thereby properly maintain substantia nigra pars compacta dopaminergic neurons and their innervation in the striatum. The demonstration of crosstalk between parkin and RET highlights the interplay in the protein network that is altered in PD and suggests potential therapeutic targets and strategies to treat PD.


Assuntos
Neurônios Dopaminérgicos/patologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Degeneração Neural/patologia , Transtornos Parkinsonianos/genética , Proteínas Proto-Oncogênicas c-ret/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Trifosfato de Adenosina/biossíntese , Animais , Ansiedade/genética , Linhagem Celular , Tamanho Celular , Progressão da Doença , Comportamento Exploratório , Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mitocôndrias/patologia , NF-kappa B/fisiologia , Transtornos Parkinsonianos/patologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-ret/deficiência , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Recombinantes de Fusão/metabolismo , Teste de Desempenho do Rota-Rod , Transdução de Sinais , Substância Negra/patologia , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/genética
8.
Neurobiol Aging ; 36(3): 1569-76, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25577412

RESUMO

Glial cell line-derived neurotrophic factor (GDNF) helps protect dopaminergic neurons in the nigrostriatal tract. Although the cause of nigrostriatal degeneration is unknown, one theory is that excess glutamate from the subthalamic nucleus results in excitotoxic events in the substantia nigra (SN). Because dopaminergic degeneration is accompanied by a reduction in GDNF, we examined glutamate neurotransmission in the SN using a Gdnf heterozygous mouse model (Gdnf(+/-)) at 8 and 12 months of age. At 8 months, Gdnf(+/-) mice have greater glutamate release and higher basal glutamate levels, which precede the SN dopaminergic degeneration observed at 12 months of age. However, at 12 months, Gdnf(+/-) mice have lower basal levels of glutamate and less glutamate release than wild-type mice. Also at 8 months, Gdnf(+/-) mice have lower levels of glutamate transporter-1 and greater glial fibrillary acidic protein levels in the SN compared with wild-type mice, differences that increase with age. These data suggest that reduced levels of GDNF induce excess glutamate release and dysregulation of glutamate transporter-1, causing excitotoxicity in the SN that precedes dopaminergic degeneration.


Assuntos
Envelhecimento/fisiologia , Transportador 2 de Aminoácido Excitatório/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Ácido Glutâmico/metabolismo , Degeneração Neural/etiologia , Degeneração Neural/patologia , Substância Negra/patologia , Substância Negra/fisiopatologia , Transmissão Sináptica/genética , Transmissão Sináptica/fisiologia , Envelhecimento/genética , Envelhecimento/patologia , Animais , Neurônios Dopaminérgicos/patologia , Feminino , Heterozigoto , Masculino , Camundongos Endogâmicos C57BL , Núcleo Subtalâmico/metabolismo
9.
Addict Biol ; 20(4): 629-42, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24801661

RESUMO

Moderate social consumption of alcohol is common; however, only a small percentage of individuals transit from social to excessive, uncontrolled alcohol drinking. This suggests the existence of protective mechanisms that prevent the development of alcohol addiction. Here, we tested the hypothesis that the glial cell line-derived neurotrophic factor (GDNF) in the mesolimbic system [e.g. the nucleus accumbens (Acb) and ventral tegmental area (VTA)] is part of such a mechanism. We found that GDNF knockdown, by infecting rat Acb neurons with a small hairpin RNA (shRNA) targeting the GDNF gene, produced a rapid escalation to excessive alcohol consumption and enhanced relapse to alcohol drinking. Conversely, viral-mediated overexpression of the growth factor in the mesolimbic system blocked the escalation from moderate to excessive alcohol drinking. To access the mechanism underlying GDNF's actions, we measured the firing rate of dopaminergic (DAergic) neurons in the VTA after a history of excessive alcohol intake with or without elevating GDNF levels. We found that the spontaneous firing rate of DAergic neurons in the VTA was reduced during alcohol withdrawal and that GDNF reversed this alcohol-induced DA deficiency. Together, our results suggest that endogenous GDNF in the mesolimbic system controls the transition from moderate to excessive alcohol drinking and relapse via reversal of alcohol-dependent neuro-adaptations in DAergic VTA neurons.


Assuntos
Consumo de Bebidas Alcoólicas/fisiopatologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Sistema Límbico/fisiologia , Núcleo Accumbens/fisiologia , Área Tegmentar Ventral/fisiologia , Adaptação Fisiológica/fisiologia , Animais , Condicionamento Operante , Neurônios Dopaminérgicos/fisiologia , Regulação para Baixo/fisiologia , Técnicas de Silenciamento de Genes , Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Masculino , Ratos Long-Evans , Recidiva , Autoadministração , Regulação para Cima/fisiologia
10.
J Neurosci Methods ; 231: 31-7, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24129039

RESUMO

BACKGROUND: Three-dimensional (3D) motion analysis is established in investigating, human pathological motion. In the field of gait, its use results in the objective identification of primary, and secondary causes of deviations, many current interventions are the result of pre- and post-testing, and it was shown recently that it can result in decreased number of surgeries and overall cost of care. Consequently, recent attempts have implemented 3D motion analysis using rat models to study, parkinsonism. However, to-date, a 3D user friendly analytical approach using rodent models to, identify etiologies of age-related motor impairment and accompanying pathologies has not been, implemented. NEW METHOD: We have developed and presented all aspects of a 3D, three body-segment rodent model, to analyze motions of the lower, upper and head segments between rodents of parkinsonism-type and, normal aging during free walking. Our model does not require transformation matrices to describe the, position of each body-segment. Because body-segment positions are not considered to consist of three, rotations about the laboratory axes, the rotations are not sequence dependent. RESULTS: Each body-segment demonstrated distinct 3D movement patterns. The parkinsonism-type, genotype walked slower with less range of motion, similarly to patients with parkinsonism. COMPARISON WITH EXISTING METHODS: This is the first model considering the rodent's body as three, distinct segments. To the best of our knowledge, it is the first model to ever consider and report the 3D, head motion patterns. CONCLUSIONS: This novel approach will allow unbiased analysis of spontaneous locomotion in mouse, models of parkinsonism or normal aging.


Assuntos
Envelhecimento/fisiologia , Modelos Biológicos , Doenças Neurodegenerativas/fisiopatologia , Transtornos Parkinsonianos/fisiopatologia , Caminhada/fisiologia , Actigrafia/métodos , Algoritmos , Animais , Fenômenos Biomecânicos , Modelos Animais de Doenças , Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Cabeça/fisiopatologia , Camundongos , Camundongos Transgênicos
11.
Pharmacol Biochem Behav ; 104: 10-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23290934

RESUMO

Glial cell-line derived neurotrophic factor (GDNF) has been established as a growth factor for the survival and maintenance of dopamine (DA) neurons. In phase I clinical trials, GDNF treatment in Parkinson's disease patients led to improved motor function and GDNF has been found to be down regulated in Parkinson's disease patients. Studies using GDNF heterozygous (Gdnf(+/-)) mice have demonstrated that a partial reduction of GDNF leads to an age-related accelerated decline in nigrostriatal DA system- and motor-function and increased neuro-inflammation and oxidative stress in the substantia nigra (SN). Therefore, the purpose of the current studies was to determine if GDNF replacement restores motor function and functional markers within the nigrostriatal DA system in middle-aged Gdnf(+/-) mice. At 11months of age, male Gdnf(+/-) and wildtype (WT) mice underwent bilateral intra-striatal injections of GDNF (10µg) or vehicle. Locomotor activity was assessed weekly 1-4weeks after treatment. Four weeks after treatment, their brains were processed for analysis of GDNF levels and various DAergic and oxidative stress markers. An intrastriatal injection of GDNF increased motor activity in Gdnf(+/-) mice to levels comparable to WT mice (1week after injection) and this effect was maintained through the 4-week time point. This increase in locomotion was accompanied by a 40% increase in striatal GDNF protein levels and SN GDNF expression in Gdnf(+/-) mice. Additionally, GDNF treatment significantly increased the number of tyrosine hydroxylase (TH)-positive neurons in the SN of middle-aged Gdnf(+/-) mice, but not WT mice, which was coupled with reduced oxidative stress in the SN. These studies further support that long-term changes related to the dysfunction of the nigrostriatal pathway are influenced by GDNF expression and add that this dysfunction appears to be responsive to GDNF treatment. Additionally, these studies suggest that long-term GDNF depletion alters the biological and behavioral responses to GDNF treatment.


Assuntos
Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Animais , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/patologia , Corpo Estriado/fisiopatologia , Ciclo-Oxigenase 2/metabolismo , Dopamina/fisiologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Neurônios Dopaminérgicos/fisiologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Substância Negra/efeitos dos fármacos , Substância Negra/patologia , Substância Negra/fisiopatologia , Superóxido Dismutase/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
12.
Neuron ; 75(6): 1051-66, 2012 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-22998873

RESUMO

The Neurotrophic factor gdnf plays diverse developmental roles, supporting survival and also acting as a chemoattractant for axon and cell migration. We report that in the developing spinal cord, a focal source of gdnf is present in the floor plate (FP) where commissural axons cross the midline. Gdnf has no direct guidance properties but switches on the responsiveness of crossing commissural growth cones to the midline repellent Semaphorin3B by suppressing calpain-mediated processing of the Sema3B signaling coreceptor Plexin-A1. Analysis of single and double mutant mouse models indicates that although gdnf is the principal trigger of Sema3B midline repulsion, it acts with another FP cue, NrCAM. Finally, genetic and in vitro experiments provide evidence that this gdnf effect is RET independent and mediated by NCAM/GFRα1 signaling. This study identifies a regulator of midline crossing and reveals interplays between Semaphorin and gdnf signaling during axon guidance.


Assuntos
Axônios/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Moléculas de Adesão de Célula Nervosa/metabolismo , Neurônios/citologia , Semaforinas/metabolismo , Análise de Variância , Animais , Axônios/efeitos dos fármacos , Padronização Corporal/genética , Calpaína/metabolismo , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Células Cultivadas , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Humanos , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Moléculas de Adesão de Célula Nervosa/genética , Neurônios/efeitos dos fármacos , Receptores de Superfície Celular/metabolismo , Semaforinas/genética , Medula Espinal/citologia , Medula Espinal/embriologia , Transfecção
13.
BMC Neurosci ; 13: 92, 2012 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-22863354

RESUMO

BACKGROUND: During development neural crest derived Schwann Cell (SC) precursors migrate to nerve trunks and populate nascent nerves. Axonal ensheathment by SC is a prerequisite for normal nerve function and the integrity of myelinated as well as nonmyelinated axons. To provide adequate support functions, SC colonize entire nerves. One important prerequisite for this is their migration into distal axonal regions. RESULTS: Here, we studied the role of Glial cell line derived neurotrophic factor (GDNF), a TGF-beta related growth factor, for SC migration. To this end we used a superior cervical ganglion (SCG) explant-SC migration assay, GDNF null mutant mouse embryos and a chemical inhibitor for GDNF signaling in combination with time-lapse imaging. We found that GDNF signaling is dispensable for SC migration along murine embryonic sympathetic axons. Furthermore, in vivo analyzes revealed that SC migration along the sciatic nerve is also not dependent on GDNF. CONCLUSIONS: In contrast to previous in vitro findings in the sciatic nerve and a SC precursor cell line, our results clearly indicate that GDNF is dispensable for embryonic SC migration. This is demonstrated for the sympathetic nervous system and also for the sciatic nerve in mouse.


Assuntos
Axônios/fisiologia , Movimento Celular/fisiologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Células de Schwann/fisiologia , Nervo Isquiático/citologia , Transdução de Sinais/genética , Animais , Afidicolina/farmacologia , Axônios/efeitos dos fármacos , Linhagem Celular Transformada , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Feminino , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Histonas/metabolismo , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Fator de Crescimento Neural/farmacologia , Gravidez , Ratos , Proteínas S100/metabolismo , Células de Schwann/efeitos dos fármacos , Nervo Isquiático/fisiologia , Gânglio Cervical Superior/citologia , Fatores de Tempo , Imagem com Lapso de Tempo , Tirosina 3-Mono-Oxigenase/metabolismo
14.
FASEB J ; 26(7): 3075-83, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22499581

RESUMO

A major therapeutic target for Parkinson's disease (PD) is providing increased glial-derived neurotrophic factor (GDNF) to dopaminergic neurons. We tested the hypothesis that innate immune activation increases astrocyte GDNF production and that this is regulated by specific eicosanoid receptors. Innate immune-activated primary murine astrocytes were assayed for GDNF expression and secretion. Controls were agent vehicle exposure and wild-type mice. Rank order for up to 10-fold selectively increased GDNF expression was activators of TLR3 > TLR2 or TLR4 > TLR9. TLR3 activator-stimulated GDNF expression was selectively JNK-dependent, followed cyclooxygenase (COX)-2, was coincident with membranous PGE(2) synthase, and was not significantly altered by a nonspecific COX- or a COX-2-selective inhibitor. Specific eicosanoid receptors had opposing effects on TLR3 activator-induced GDNF expression: ∼60% enhancement by blocking or ablating of PGE(2) receptor subtype 1 (EP1), ∼30% enhancement by activating PGF(2α) receptor or thromboxane receptor, or ∼15% enhancement by activating EP4. These results demonstrate functionally antagonistic eicosanoid receptor subtype regulation of innate immunity-induced astrocyte GDNF expression and suggest that selective inhibition of EP1 signaling might be a means to augment astrocyte GDNF secretion in the context of innate immune activation in diseased regions of brain in PD.


Assuntos
Astrócitos/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Receptores Eicosanoides/metabolismo , Receptores Toll-Like/metabolismo , Animais , Astrócitos/imunologia , Sequência de Bases , Células Cultivadas , Primers do DNA/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Receptores Eicosanoides/classificação , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 3 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Receptor Toll-Like 9/metabolismo
15.
Neuroscience ; 171(4): 1357-66, 2010 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-20933580

RESUMO

Glial cell line-derived neurotrophic factor (GDNF) is a potent factor for the ventral mesencephalic dopamine neurons. However, studies on the Gdnf gene deleted (Gdnf(-/-)) mouse have been limited to fetal tissue since these mice die prematurely. To evaluate long-term effects of Gdnf gene deletion, this study involves co-grafts of ventral mesencephalon (VM) and lateral ganglionic eminence (LGE) derived from different Gdnf genotypes. The VM/LGE co-grafts were evaluated at 3, 6, and 12 months for tyrosine hydroxylase (TH) -positive cell survival and nerve fiber formation in the LGE co-transplant, visualized by dopamine- and cyclic AMP-regulated phosphoprotein relative molecular mass 32,000 (DARPP-32) -immunoreactivity. Cell counts revealed no difference in TH-positive neurons between Gdnf genotypes at 3 months postgrafting. At 6 months, a significant reduction in cell number was observed in the Gdnf(-/-) grafts. In fact, in the majority of the Gdnf(-/-) VM/LGE transplant had degenerated. At 12 months, a reduction in cell number was seen in both Gdnf(-/-) and Gdnf(+/-) compared to wild type transplants. In the Gdnf(-/-) grafts, TH-negative inclusion-like structures were present in the cytoplasm of the TH-positive neurons at 3 months. These structures were also found in the Gdnf(+/-) transplants at 12 months, but not in Gdnf(+/+) controls at any time point. In Gdnf(+/+) grafts, TH-positive nerve fiber innervation of the striatal co-grafts was dense and patchy and overlapped with clusters of DARPP-32-positive neurons. This overlap did mismatch in the Gdnf(+/-) grafts, while the TH-positive innervation was sparse in the Gdnf(-/-) transplants and the DARPP-32-positive neurons were widespread distributed. In conclusion, GDNF is essential for long-term maintenance of both the VM TH-positive neurons and for the striatal tissue, and appears crucial for generation of a proper organization of the striatum.


Assuntos
Corpo Estriado/fisiologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Substância Negra/fisiologia , Aldeído Desidrogenase/metabolismo , Família Aldeído Desidrogenase 1 , Análise de Variância , Animais , Vasos Sanguíneos/metabolismo , Sobrevivência Celular , Corpo Estriado/cirurgia , Embrião de Mamíferos , Transplante de Tecido Fetal/métodos , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Isoenzimas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/fisiologia , Fibras Nervosas/metabolismo , Vias Neurais/fisiologia , Retinal Desidrogenase , Substância Negra/cirurgia , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/metabolismo , alfa-Sinucleína/metabolismo
16.
J Neural Transm (Vienna) ; 117(6): 719-27, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20458508

RESUMO

Loss of dopaminergic neurons in the substantia nigra pars compacta and the resulting decrease in striatal dopamine levels are the hallmarks of Parkinson's disease. Tgfbeta and Gdnf have been identified as neurotrophic factors for dopaminergic midbrain neurons in vivo and in vitro. Haploinsufficiency for either Tgfbeta or Gdnf led to dopaminergic deficits. In this study we therefore analyzed the nigrostriatal system of aged Tgfbeta2 (+/-)/Gdnf (+/-) double-heterozygous mice. Unexpectedly, we found no morphological changes in the nigrostriatal system as compared with age-matched wild-type mice. There were no significant differences in the number of TH-positive midbrain neurons and no changes in the optical density of TH immunoreactivity in striata of Tgfbeta2 (+/-)/Gdnf (+/-) double-heterozygous mice. Moreover, we found no significant differences in the striatal levels of dopamine and its metabolites dihydroxyphenylacetic acid and homovanillic acid. Our results indicate that a combined haploinsufficiency for Tgfbeta2 and Gdnf has no impact on the function and the survival of midbrain DA neurons under normal aging conditions.


Assuntos
Envelhecimento , Corpo Estriado/patologia , Regulação da Expressão Gênica/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Substância Negra/patologia , Fator de Crescimento Transformador beta2/genética , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Análise de Variância , Animais , Cromatografia Líquida de Alta Pressão/métodos , Dopamina/metabolismo , Eletroquímica/métodos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Ácido Homovanílico/metabolismo , Camundongos , Camundongos Transgênicos , Vias Neurais/patologia , RNA Mensageiro/metabolismo , Fator de Crescimento Transformador beta2/deficiência , Tirosina 3-Mono-Oxigenase/metabolismo
17.
Hum Gene Ther ; 20(7): 715-27, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19374591

RESUMO

Deprivation of neurotrophic factors contributes to the pathogenesis of diabetic neuropathy. However, the role of glial cell-derived neurotrophic factor (GDNF) in the pathogenesis of diabetic neuropathy remains unclear. The present study evaluated the pathogenic role of GDNF deficiency and the therapeutic potential of GDNF gene transfer for diabetic neuropathy. After injection of streptozotocin (STZ) for 2 weeks, diabetic rats displayed significant alteration in electrophysiological parameters, which was associated with structural changes and defective myelination in the sciatic nerves. The early diabetic neuropathy was accompanied by attenuation of the GDNF/GFRalpha1/Akt signaling cascade and depletion of sensory neuropeptides in the peripheral nerves. After detection of neuropathy, intramuscular GDNF gene transfer reversed the deficiency of GDNF/Akt signaling in the sciatic nerve and improved the neurological functions of diabetic rats. Moreover, GDNF gene delivery alleviated the axonal demyelination and restored the sensory neuropeptide levels in the sciatic nerve of diabetic rats. In summary, peripheral GDNF gene delivery ameliorates the diabetes-induced downregulation of the GDNF signaling complex in the peripheral nervous system and holds promises for treatment of diabetic neuropathy.


Assuntos
Neuropatias Diabéticas/patologia , Neuropatias Diabéticas/terapia , Técnicas de Transferência de Genes , Terapia Genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/uso terapêutico , Nervo Isquiático/metabolismo , Animais , Axônios/patologia , Axônios/ultraestrutura , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Neuropatias Diabéticas/complicações , Neuropatias Diabéticas/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/administração & dosagem , Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Hiperglicemia/complicações , Hiperglicemia/patologia , Injeções Intramusculares , Bainha de Mielina/patologia , Bainha de Mielina/ultraestrutura , Ratos , Nervo Isquiático/ultraestrutura , Transdução de Sinais , Estreptozocina , Substância P/metabolismo
18.
J Neurosci ; 28(30): 7467-75, 2008 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-18650325

RESUMO

Neurotrophic factors, including glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF), promote survival of midbrain dopaminergic neurons, but the death pathways activated in the dopaminergic neurons by deprivation of these factors are poorly studied. We show here that deprivation of GDNF or BDNF triggers a novel mitochondria-independent death pathway in the cultured embryonic dopaminergic neurons: cytochrome c was not released from the mitochondria to cytosol, proapoptotic protein Bax was not activated, and overexpressed Bcl-xL did not block the death. Caspases were critically required, because the death was completely blocked by caspase inhibitor BAF [boc-aspartyl(OMe)-fluoromethylketone] and overexpression of dominant-negative mutants of caspase-9, -3, and -7 significantly blocked the death. Also, the death receptor pathway was involved, because blockage of caspase-8 or FADD (Fas-associated protein with death domain), an adapter required for caspase-8 activation, inhibited death induced by GDNF or BDNF deprivation. Ligation of Fas by agonistic anti-Fas antibody induced apoptosis in the GDNF- or BDNF-maintained neurons, and inhibition of Fas by Fas-Fc chimera blocked the death of GDNF- or BDNF-deprived neurons, whereas FAIM(L) (long isoform of Fas apoptosis inhibitory molecule) could control the activity of Fas in the dopaminergic neurons.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/deficiência , Caspases/metabolismo , Dopamina/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Mesencéfalo/citologia , Mitocôndrias/fisiologia , Neurônios/metabolismo , Receptores de Morte Celular/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Antígenos Nucleares/metabolismo , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/fisiologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Homeodomínio/metabolismo , Imunoprecipitação , Autoantígeno Ku , Camundongos , Estaurosporina/farmacologia , Fatores de Transcrição/metabolismo , Transfecção/métodos , Tirosina 3-Mono-Oxigenase/metabolismo , Proteína bcl-X/metabolismo
19.
Glia ; 56(13): 1428-37, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18551627

RESUMO

The phenotypic development of satellite cells in mouse sympathetic ganglia was examined by localizing the transcription factors, Sox10 and Phox2b, the neuronal marker, tyrosine hydroxylase (TH), and brain-derived fatty acid binding protein (B-FABP), which identifies glial precursors and mature glia. In E10.5 mice, most cells in the sympathetic chain expressed both Sox10 and Phox2b, with a minority of cells expressing Sox10 only or Phox2b only. In E11.5 mice, the majority of cells expressed Sox10 only or Phox2b only. B-FABP was colocalized with Sox10 in satellite glial precursors, which were located on the periphery of the ganglion. There was no overlap between B-FABP and Phox2b or B-FABP and TH. During subsequent development, the number of B-FABP+ cells increased and they became more common deep within the ganglion. In E12.5 and E18.5 mice, there was no overlap between Sox10 and Phox2b, and 98% of Sox10 cells were also B-FABP+. Satellite glial precursors in E11.5-E15.5 mice also expressed the GDNF-binding molecule, GFRalpha1. B-FABP immunoreactive cells did not express Ret or NCAM, two potential signaling molecules for GDNF/GFRalpha1. In E12.5 and E18.5 mice lacking GFRalpha1 or GDNF, the development of B-FABP immunoreactive satellite cells was normal, and hence neither GDNF or GFRalpha1 are essential for the development of satellite glia in sympathetic ganglia.


Assuntos
Gânglios Simpáticos/citologia , Gânglios Simpáticos/crescimento & desenvolvimento , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Neuroglia/citologia , Neuroglia/fisiologia , Animais , Feminino , Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Gravidez
20.
Nat Neurosci ; 11(7): 755-61, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18536709

RESUMO

GDNF is a potent neurotrophic factor that protects catecholaminergic neurons from toxic damage and induces fiber outgrowth. However, the actual role of endogenous GDNF in the normal adult brain is unknown, even though GDNF-based therapies are considered promising for neurodegenerative disorders. We have generated a conditional GDNF-null mouse to suppress GDNF expression in adulthood, hence avoiding the developmental compensatory modifications masking its true physiologic action. After Gdnf ablation, mice showed a progressive hypokinesia and a selective decrease of brain tyrosine hydroxylase (Th) mRNA, accompanied by pronounced catecholaminergic cell death, affecting most notably the locus coeruleus, which practically disappears; the substantia nigra; and the ventral tegmental area. These data unequivocally demonstrate that GDNF is indispensable for adult catecholaminergic neuron survival and also show that, under physiologic conditions, downregulation of a single trophic factor can produce massive neuronal death.


Assuntos
Encéfalo/citologia , Catecolaminas/metabolismo , Regulação da Expressão Gênica/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Neurônios/metabolismo , Animais , Antineoplásicos Hormonais/toxicidade , Comportamento Animal/efeitos dos fármacos , Contagem de Células/métodos , Sobrevivência Celular/genética , Colina O-Acetiltransferase/metabolismo , Ensaio de Imunoadsorção Enzimática/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Glutamato Descarboxilase/metabolismo , Hipocinesia/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fosfopiruvato Hidratase/metabolismo , Tamoxifeno/toxicidade , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...