Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Int J Mol Sci ; 23(19)2022 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-36232361

RESUMO

Synucleinopathies are a group of neurodegenerative disorders caused by the accumulation of toxic species of α-synuclein. The common clinical features are chronic progressive decline of motor, cognitive, behavioral, and autonomic functions. They include Parkinson's disease, dementia with Lewy body, and multiple system atrophy. Their etiology has not been clarified and multiple pathogenic factors include oxidative stress, mitochondrial dysfunction, impaired protein degradation systems, and neuroinflammation. Current available therapy cannot prevent progressive neurodegeneration and "disease-modifying or neuroprotective" therapy has been proposed. This paper presents the molecular mechanisms of neuroprotection by the inhibitors of type B monoamine oxidase, rasagiline and selegiline. They prevent mitochondrial apoptosis, induce anti-apoptotic Bcl-2 protein family, and pro-survival brain- and glial cell line-derived neurotrophic factors. They also prevent toxic oligomerization and aggregation of α-synuclein. Monoamine oxidase is involved in neurodegeneration and neuroprotection, independently of the catalytic activity. Type A monoamine oxidases mediates rasagiline-activated signaling pathways to induce neuroprotective genes in neuronal cells. Multi-targeting propargylamine derivatives have been developed for therapy in various neurodegenerative diseases. Preclinical studies have presented neuroprotection of rasagiline and selegiline, but beneficial effects have been scarcely presented. Strategy to improve clinical trials is discussed to achieve disease-modification in synucleinopathies.


Assuntos
Doenças Neurodegenerativas , Fármacos Neuroprotetores , Sinucleinopatias , Fatores Neurotróficos Derivados de Linhagem de Célula Glial , Humanos , Indanos/farmacologia , Indanos/uso terapêutico , Monoaminoxidase/metabolismo , Inibidores da Monoaminoxidase/farmacologia , Inibidores da Monoaminoxidase/uso terapêutico , Doenças Neurodegenerativas/tratamento farmacológico , Neuroproteção , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Selegilina/farmacologia , alfa-Sinucleína
2.
Development ; 147(21)2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-32994173

RESUMO

Appropriately balanced RET signaling is of crucial importance during embryonic neural crest cell migration, proliferation and differentiation. RET deficiency, for example, leads to intestinal aganglionosis (Hirschsprung disease), whereas overactive RET can lead to multiple endocrine neoplasia (MEN) syndromes. Some RET mutations are associated with both intestinal aganglionosis and MEN-associated tumors. This seemingly paradoxical occurrence has led to speculation of a 'Janus mutation' in RET that causes overactivation or impairment of RET activity depending on the cellular context. Using an intestinal catenary culture system to test the effects of GDNF-mediated RET activation, we demonstrate the concurrent development of distal colonic aganglionosis and intestinal ganglioneuromas. Interestingly, the tumors induced by GDNF stimulation contain enteric neuronal progenitors capable of reconstituting an enteric nervous system when transplanted into a normal developmental environment. These results suggest that a Janus mutation may not be required to explain co-existing Hirschsprung disease and MEN-associated tumors, but rather that RET overstimulation alone is enough to cause both phenotypes. The results also suggest that reprogramming tumor cells toward non-pathological fates may represent a possible therapeutic avenue for MEN-associated neoplasms.


Assuntos
Ganglioneuroma/patologia , Doença de Hirschsprung/patologia , Intestinos/patologia , Proteínas Proto-Oncogênicas c-ret/metabolismo , Animais , Agregação Celular , Diferenciação Celular , Embrião de Galinha , Galinhas , Sistema Nervoso Entérico/patologia , Ganglioneuroma/metabolismo , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/metabolismo , Doença de Hirschsprung/metabolismo , Camundongos Endogâmicos C57BL , Crista Neural/patologia , Neurônios/metabolismo , Neurônios/patologia , Nervo Vago/patologia
3.
Mol Pain ; 16: 1744806920950866, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32811276

RESUMO

The glial cell line-derived neurotrophic factor (GDNF) family ligands (GFLs) alleviate symptoms of experimental neuropathy, protect and stimulate regeneration of sensory neurons in animal models of neuropathic pain, and restore their functional activity. However, clinical development of GFL proteins is complicated by their poor pharmacokinetic properties and multiple effects mediated by several receptors. Previously, we have identified a small molecule that selectively activates the major signal transduction unit of the GFL receptor complex, receptor tyrosine kinase RET, as an alternative to GFLs, for the treatment of neuropathic pain. We then introduced a series of chemical changes to improve the biological activity of these compounds and tested an optimized compound named BT44 in a panel of biological assays. BT44 efficiently and selectively stimulated the GFL receptor RET and activated the intracellular mitogene-activated protein kinase/extracellular signal-regulated kinase pathway in immortalized cells. In cultured sensory neurons, BT44 stimulated neurite outgrowth with an efficacy comparable to that of GFLs. BT44 alleviated mechanical hypersensitivity in surgery- and diabetes-induced rat models of neuropathic pain. In addition, BT44 normalized, to a certain degree, the expression of nociception-related neuronal markers which were altered by spinal nerve ligation, the neuropathy model used in this study. Our results suggest that the GFL mimetic BT44 is a promising new lead for the development of novel disease-modifying agents for the treatment of neuropathy and neuropathic pain.


Assuntos
Biomimética/métodos , Neuralgia/tratamento farmacológico , Proteínas Proto-Oncogênicas c-ret/agonistas , Proteínas Proto-Oncogênicas c-ret/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Nervos Espinhais/efeitos dos fármacos , Animais , Escala de Avaliação Comportamental , Linhagem Celular , Neuropatias Diabéticas/tratamento farmacológico , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fatores Neurotróficos Derivados de Linhagem de Célula Glial , Imuno-Histoquímica , Masculino , Proteínas do Tecido Nervoso/metabolismo , Neuralgia/metabolismo , Nociceptividade/efeitos dos fármacos , Fosforilação , Ratos , Ratos Wistar , Células Receptoras Sensoriais/metabolismo , Nervos Espinhais/lesões
4.
Brain Pathol ; 29(6): 813-825, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31033033

RESUMO

Growth factors can facilitate hippocampus-based learning and memory and are potential targets for treatment of cognitive dysfunction via their neuroprotective and neurorestorative effects. Dementia is common in Parkinson's disease (PD), but treatment options are limited. We aimed to determine if levels of growth factors are altered in the hippocampus of patients with PD, and if such alterations are associated with PD pathology. Enzyme-linked immunosorbent assays were used to quantify seven growth factors in fresh frozen hippocampus from 10 PD and nine age-matched control brains. Western blotting and immunohistochemistry were used to explore cellular and inflammatory changes that may be associated with growth factor alterations. In the PD hippocampus, protein levels of glial cell line-derived neurotrophic factor were significantly decreased, despite no evidence of neuronal loss. In contrast, protein levels of fibroblast growth factor 2 and cerebral dopamine neurotrophic factor were significantly increased in PD compared to controls. Levels of the growth factors epidermal growth factor, heparin-binding epidermal growth factor, brain-derived neurotrophic factor and mesencephalic astrocyte-derived neurotrophic factor did not differ between groups. Our data demonstrate changes in specific growth factors in the hippocampus of the PD brain, which potentially represent targets for modification to help attenuate cognitive decline in PD. These data also suggest that multiple growth factors and direction of change needs to be considered when approaching growth factors as a potential treatment for cognitive decline.


Assuntos
Hipocampo/metabolismo , Fatores de Crescimento Neural/metabolismo , Doença de Parkinson/metabolismo , Idoso , Idoso de 80 Anos ou mais , Encéfalo/patologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Corpo Estriado/patologia , Dopamina/metabolismo , Feminino , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/metabolismo , Hipocampo/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/análise , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Neuroglia/metabolismo , Substância Negra/patologia
5.
Mol Reprod Dev ; 85(8-9): 720-728, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29969526

RESUMO

Spermatogonial stem cells (SSCs), a unique population of male germ cells with self-renewal ability, are the foundation for maintenance of spermatogenesis throughout the life of the male. Although many regulatory molecules essential for SSC self-renewal have been identified, the fundamental mechanism underlying how SSCs acquire and maintain their self-renewal activity remains largely to be elucidated. In recent years, many types of noncoding RNAs (ncRNAs) have been suggested to regulate the SSC self-renewal through multiple ways, indicating ncRNAs play crucial roles in SSC self-renewal. In this paper, we mainly focus on four types of ncRNAs including microRNA, long ncRNA, piwi-interacting RNA, as well as circular RNAs, and reviewed their potential roles in SSC self-renewal that discovered recently to help us gain a better understanding of molecular mechanisms by which ncRNAs perform their function in regulating SSC self-renewal.


Assuntos
Células-Tronco Germinativas Adultas/fisiologia , Proliferação de Células/fisiologia , RNA não Traduzido/fisiologia , Espermatogênese/fisiologia , Animais , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/fisiologia , Humanos , Masculino , Mamíferos , Camundongos
6.
J Surg Res ; 219: 214-221, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29078884

RESUMO

BACKGROUND: The opioid epidemic is a growing concern, and emerging evidence suggests that morphine use may be associated with sepsis. Enteric glial cells (EGCs) are the most numerous cell type in the enteric nervous system and regulate gastrointestinal function through the production of trophic factors, including glial-derived neurotrophic factor (GDNF). We sought to determine the effect of morphine on enteric glia and hypothesized that morphine contributes to EGC dysfunction and increased gut permeability. MATERIALS AND METHODS: Rat intestinal epithelial cells (IECs) and EGC lines were purchased from ATCC. Immunocytochemistry was used to evaluate the impact of EGCs on IEC barrier proteins and detect the µ-opioid receptor. Co-culture assays were used to determine the effect of EGCs, GDNF, and morphine on barrier integrity. Quantitative polymerase chain reaction and western blotting were performed to determine the impact of morphine in GDNF production. Transepithelial resistance of IEC-6 cell monolayers was measured in the presence of EGC-conditioned media (EGC-CM) and morphine treated EGC-CM using electrical cell impedance sensing. RESULTS: EGC-CM enhanced tight junction organization in IECs. IEC barrier integrity was enhanced when co-cultured with unstimulated EGCs or with GDNF alone; this barrier protective effect was lost with morphine-treated EGCs. GDNF RNA and protein expression were decreased by morphine treatment. Transepithelial resistance was decreased in IEC confluent monolayers when exposed to morphine-treated EGC-CM compared with control. CONCLUSIONS: Morphine compromises intestinal epithelial cell barrier function through a mechanism which appears to involve GDNF. Further studies are warranted to delineate the role of enteric glial cell function in opioid signaling and sepsis.


Assuntos
Analgésicos Opioides/efeitos adversos , Mucosa Intestinal/efeitos dos fármacos , Morfina/efeitos adversos , Neuroglia/efeitos dos fármacos , Animais , Linhagem Celular , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/metabolismo , Neuroglia/química , Neuroglia/metabolismo , Ratos , Receptores Opioides mu/análise
7.
Immunity ; 45(2): 238-9, 2016 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-27533011

RESUMO

Type 3 innate lymphoid cells (ILC3s) and enteric glia, an essential structural component of gut innervation, are well-known regulators of intestinal homeostasis. Ibiza et al. (2016) uncover a new link between commensal bacteria, enteric glial cells, and ILC3s that is required for intestinal homeostasis and defense.


Assuntos
Disbiose/genética , Microbioma Gastrointestinal/imunologia , Imunidade Inata , Intestinos/imunologia , Linfócitos/imunologia , Neuroglia/imunologia , Proteínas Proto-Oncogênicas c-ret/metabolismo , Animais , Técnicas de Silenciamento de Genes , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/metabolismo , Homeostase , Humanos , Interleucinas/metabolismo , Intestinos/inervação , Camundongos , Neuroglia/microbiologia , Proteínas Proto-Oncogênicas c-ret/genética , Simbiose
8.
J Neurosci ; 35(38): 13233-43, 2015 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-26400951

RESUMO

Glial cell line-derived neurotrophic factor (GDNF) promotes PNS development and kidney morphogenesis via a receptor complex consisting of the glycerophosphatidylinositol (GPI)-anchored, ligand binding receptor GDNF family receptor α1 (GFRα1) and the receptor tyrosine kinase Ret. Although Ret signal transduction in vitro is augmented by translocation into lipid rafts via GFRα1, the existence and importance of lipid rafts in GDNF-Ret signaling under physiologic conditions is unresolved. A knock-in mouse was produced that replaced GFRα1 with GFRα1-TM, which contains a transmembrane (TM) domain instead of the GPI anchor. GFRα1-TM still binds GDNF and promotes Ret activation but does not translocate into rafts. In Gfrα1(TM/TM) mice, GFRα1-TM is expressed, trafficked, and processed at levels identical to GFRα1. Although Gfrα1(+/TM) mice are viable, Gfrα1(TM/TM) mice display bilateral renal agenesis, lack enteric neurons in the intestines, and have motor axon guidance deficits, similar to Gfrα1(-/-) mice. Therefore, the recruitment of Ret into lipid rafts by GFRα1 is required for the physiologic functions of GDNF in vertebrates. Significance statement: Membrane microdomains known as lipid rafts have been proposed to be unique subdomains in the plasma membrane that are critical for the signaling functions of multiple receptor complexes. Their existence and physiologic relevance has been debated. Based on in vitro studies, lipid rafts have been reported to be necessary for the function of the Glial cell line-derived neurotrophic factor (GDNF) family of neurotrophic factors. The receptor for GDNF comprises the lipid raft-resident, glycerophosphatidylinositol-anchored receptor GDNF family receptor α1 (GFRα1) and the receptor tyrosine kinase Ret. Here we demonstrate, using a knock-in mouse model in which GFRα1 is no longer located in lipid rafts, that the developmental functions of GDNF in the periphery require the translocation of the GDNF receptor complex into lipid rafts.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/metabolismo , Microdomínios da Membrana/fisiologia , Morfogênese/fisiologia , Neurônios/citologia , Acetilcolinesterase/metabolismo , Animais , Células Cultivadas , Dipeptídeos/farmacologia , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Estrenos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/genética , Humanos , Ácidos Hidroxâmicos/farmacologia , Microdomínios da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Morfogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Pirrolidinonas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Medula Espinal/citologia , Gânglio Cervical Superior/citologia , Tirosina 3-Mono-Oxigenase/metabolismo
9.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 33(1): 16-20, 2015 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-25872292

RESUMO

OBJECTIVE: This research aims to study the changes in pain threshold and glial cell line-derived neurotrophic factor (GDNF) in a Sprague Dawley (SD) rat model oftrigeminal neuralgia. METHODS: A total of 36 male SD rats were randomly divided into three groups: operative, sham-operative, and control. In the operative group, a chronic constriction injury (CCI) was caused by placing loose chromic gut ligatures around the right infraorbital nerve (ION). In the sham-operative group, the right ION was subjected to the same procedure, but without ligation. In the control group, the right ION was not subjected to any treatment. The pain thresholds of the three groups were recorded at different times after the operation. The GDNF expression in each group was analyzed via immunohistochemical staining. RESULTS: An allodynia to mechanical stimulation in the region of the ligated ION was observed starting on the 2nd week after operation. Pain thresholds started to increase gradually from the 6th week and returned to the original level at the 10th to 12th week after operation. Cells that expressed the GDNF markedly increased in number in the operative group with changes observed at different times. CONCLUSION: We use chronic constriction injury to the infraorbital nerve (CCI-ION) to establish a trigeminal neuralgia-like animal model in SD rats. GDNF may play a role in regulating pain by promoting the restoration and regeneration of nerve fibers.


Assuntos
Limiar da Dor , Neuralgia do Trigêmeo , Animais , Constrição , Modelos Animais de Doenças , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Fatores Neurotróficos Derivados de Linhagem de Célula Glial , Hiperalgesia , Masculino , Ratos , Ratos Sprague-Dawley
10.
Drug Des Devel Ther ; 9: 1889-96, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25848224

RESUMO

BACKGROUND: Glial cell line-derived neurotrophic factor (GDNF) is a small protein that potently promotes the survival of many types of neurons. Detection of GDNF is vital to monitoring the survival of sympathetic and sensory neurons. However, the specific method for GDNF detection is also un-discovered. The purpose of this study is to explore the method for protein detection of GDNF. METHODS: A novel visual detection method based on a molecular translator and isothermal strand-displacement polymerization reaction (ISDPR) has been proposed for the detection of GDNF. In this study, a molecular translator was employed to convert the input protein to output deoxyribonucleic acid signal, which was further amplified by ISDPR. The product of ISDPR was detected by a lateral flow biosensor within 30 minutes. RESULTS: This novel visual detection method based on a molecular translator and ISDPR has very high sensitivity and selectivity, with a dynamic response ranging from 1 pg/mL to 10 ng/mL, and the detection limit was 1 pg/mL of GDNF. CONCLUSION: This novel visual detection method exhibits high sensitivity and selectivity, which is very simple and universal for GDNF detection to help disease therapy in clinical practice.


Assuntos
Técnicas Biossensoriais/métodos , DNA/análise , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/análise , Polimerização , Temperatura , DNA/química , Humanos , Sensibilidade e Especificidade
11.
Neurol Med Chir (Tokyo) ; 55(2): 113-23, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25746305

RESUMO

Regenerative medicine for Parkinson's disease (PD) is expected to develop dramatically with the advancement of biotechnology as represented by induced pluripotent stem cells. Existing therapeutic strategy for PD consists of medication using L-DOPA, surgery such as deep brain stimulation and rehabilitation. Current treatment cannot stop the progression of the disease, although there is definite therapeutic effect. True neurorestoration is strongly desired by regenerative medicine. This review article describes the historical development of regenerative medicine for PD, with a focus on fetal nigral cell transplantation and glial cell line-derived neurotrophic factor infusion. Subsequently, the current status of regenerative medicine for PD in terms of cell therapy and gene therapy are reviewed. In the end, the future direction to realize regenerative medicine for PD is discussed.


Assuntos
Doença de Parkinson/terapia , Medicina Regenerativa/métodos , Animais , Antiparkinsonianos/uso terapêutico , Terapia Baseada em Transplante de Células e Tecidos/métodos , Ensaios Clínicos como Assunto , Terapia Combinada , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/fisiologia , Descoberta de Drogas , Células-Tronco Fetais/transplante , Terapia Genética/métodos , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/uso terapêutico , Humanos , Levodopa/uso terapêutico , Doença de Parkinson/fisiopatologia , Células-Tronco Pluripotentes/transplante , Substância Negra/citologia
12.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-261146

RESUMO

<p><b>OBJECTIVE</b>This research aims to study the changes in pain threshold and glial cell line-derived neurotrophic factor (GDNF) in a Sprague Dawley (SD) rat model oftrigeminal neuralgia.</p><p><b>METHODS</b>A total of 36 male SD rats were randomly divided into three groups: operative, sham-operative, and control. In the operative group, a chronic constriction injury (CCI) was caused by placing loose chromic gut ligatures around the right infraorbital nerve (ION). In the sham-operative group, the right ION was subjected to the same procedure, but without ligation. In the control group, the right ION was not subjected to any treatment. The pain thresholds of the three groups were recorded at different times after the operation. The GDNF expression in each group was analyzed via immunohistochemical staining.</p><p><b>RESULTS</b>An allodynia to mechanical stimulation in the region of the ligated ION was observed starting on the 2nd week after operation. Pain thresholds started to increase gradually from the 6th week and returned to the original level at the 10th to 12th week after operation. Cells that expressed the GDNF markedly increased in number in the operative group with changes observed at different times.</p><p><b>CONCLUSION</b>We use chronic constriction injury to the infraorbital nerve (CCI-ION) to establish a trigeminal neuralgia-like animal model in SD rats. GDNF may play a role in regulating pain by promoting the restoration and regeneration of nerve fibers.</p>


Assuntos
Animais , Masculino , Ratos , Constrição , Modelos Animais de Doenças , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Fatores Neurotróficos Derivados de Linhagem de Célula Glial , Hiperalgesia , Limiar da Dor , Ratos Sprague-Dawley , Neuralgia do Trigêmeo
13.
Curr Diabetes Rev ; 10(3): 166-76, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24809393

RESUMO

Diabetes mellitus is a disease with a devastating impact on population. Recent data revealed that early retinal neuropathy in patients with diabetic retinopathy involved a reduced expression of brain-derived neurotrophic factor. Retinal ganglion cells (RGC) neuropathy is a progressive optic nerve neuropathy with RGC death and axonal degeneration, and it leads to blindness in the elderly population worldwide. Thus, neuroprotective therapies that rescue damaged RGCs and inhibit the progression of RGC loss and axonal degeneration are needed. This review introduces potential neuroprotective therapies using different neurotrophic factors for damaged RGC in eyes with RGC neuropathy associated diseases.


Assuntos
Cegueira/patologia , Neuropatias Diabéticas/patologia , Fatores de Crescimento Neural/farmacologia , Fármacos Neuroprotetores/farmacologia , Retina/patologia , Células Ganglionares da Retina/patologia , Idoso , Cegueira/tratamento farmacológico , Cegueira/prevenção & controle , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Neuropatias Diabéticas/tratamento farmacológico , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/metabolismo , Humanos , Células Ganglionares da Retina/efeitos dos fármacos
14.
Biochem Biophys Res Commun ; 449(3): 338-43, 2014 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-24853803

RESUMO

We examined the effects of conditioned medium from olfactory ensheathing glia (OEGCM) on the differentiation of oligodendrocytes in mixed cultures of early postnatal hippocampi. Differentiation was judged from the numerical density (ND) of cells immunoreactive to 2'3' cyclic nucleotide 3'phosphodiesterase (CNPase) and O4 antibodies. NDs increased according to inverted-U dose-response curves, particularly for CNPase+ cells (9-fold at optimal dilution) and these changes were blocked by inhibitors of ERK1, p38-MAPK, and PI3K. Our results raise the possibility that OEG secreted factor(s) may counteract demyelination induced by trauma, neurodegenerative diseases, and advanced age, and should stimulate novel methods to deliver these factors and/or potentiating chemicals.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/farmacologia , Neuroglia/metabolismo , Bulbo Olfatório/metabolismo , Oligodendroglia/efeitos dos fármacos , 2',3'-Nucleotídeo Cíclico Fosfodiesterases/metabolismo , Animais , Contagem de Células , Técnicas de Cultura de Células , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/metabolismo , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Oligodendroglia/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Ratos , Ratos Wistar , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Med Hypotheses ; 82(6): 681-7, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24661939

RESUMO

Exercise is a well known neuroprotective and neurotherapeutic strategy in animal models and humans with brain injury and cognitive dysfunction. In part, exercise induced beneficial effects relate to endothelial derived nitric oxide (eNO) production and induction of the neurotrophins; Brain Derived Neurotrophic Factor (BDNF) and Glial Derived Neurotrophic Factor (GDNF). Whole Body Periodic Acceleration (WBPA (pGz), is the motion of the supine body headward to footward in a sinusoidal fashion, at frequencies of 100-160 cycles/min, inducing pulsatile shear stress to the vascular endothelium. WBPA (pGz) increases eNO in the cardiovascular system in animal models and humans. We hypothesized that WBPA (pGz) has neuroprotective and neurotherapeutic effects due to enhancement of biological pathways that include eNOS, BDNF and GDNF. We discuss protein expression analysis of these in brain of rodents. Animal and observational human data affirm a neuroprotective and neurotherapeutic role for WBPA (pGz). These findings suggest that WBPA (pGz) in addition to its well known beneficial cardiovascular effects can be a simple non-invasive neuroprotective and neurotherapeutic strategy with far reaching health benefits.


Assuntos
Aceleração , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Encéfalo/metabolismo , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/metabolismo , Manipulações Musculoesqueléticas/métodos , Fenômenos Fisiológicos do Sistema Nervoso/fisiologia , Óxido Nítrico/metabolismo , Animais , Humanos , Camundongos , Ratos
16.
Mol Reprod Dev ; 81(3): 217-29, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24293385

RESUMO

It is well documented that oocytes from small antral follicles are less competent than those derived from large follicles, and we have previously shown that glial cell line-derived neurotrophic factor (GDNF) enhances developmental competence in oocytes from antral follicles. Exactly how GDNF effects this change and if it depends on the stage of oocyte development is currently unknown. The objective of this study was to examine the transcriptomic effects of follicle size and GDNF on the in vitro maturation of porcine oocytes. Microarray analysis uncovered differentially expressed transcripts among in vitro-matured porcine oocytes from different-size antral follicles, in the absence or presence of GDNF. Oocytes isolated from small follicles showed a lower state of maturation than those from large follicles, with several transcripts associated with meiotic arrest. Addition of GDNF to the culture media had effects that depended on the stage of the follicle from which the oocyte was isolated, with those from small follicles showing decreased expression of genes associated with acetyltransferase activity while those from large follicles showed decreased metabolic activity. In summary, our results revealed considerable differences between the transcriptomes of small- and large-follicle-derived oocytes. Furthermore, GDNF affects the developmental competence of oocytes in follicle-stage dependent manner. Thus, improving our understanding of the requirements for successful in vitro maturation of porcine oocytes will inform current reproductive technologies, with implications for the future of animal and human health.


Assuntos
Fatores Neurotróficos Derivados de Linhagem de Célula Glial/farmacologia , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Transcriptoma/efeitos dos fármacos , Animais , Blastocisto/efeitos dos fármacos , Blastocisto/metabolismo , Análise por Conglomerados , Feminino , Humanos , Técnicas de Maturação in Vitro de Oócitos , Masculino , Oócitos/metabolismo , RNA Mensageiro/análise , RNA Mensageiro/isolamento & purificação , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real , Suínos
17.
PLoS One ; 8(9): e72926, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24069165

RESUMO

Administration of mesenchymal stromal cells (MSC) improves functional outcome in the SOD1G93A mouse model of the degenerative motor neuron disorder amyotrophic lateral sclerosis (ALS) as well as in models of other neurological disorders. We have now investigated the effect of the interaction between MSC and motor neurons (derived from both non-transgenic and mutant SOD1G93A transgenic mice), NSC-34 cells and glial cells (astrocytes, microglia) (derived again from both non-transgenic and mutant SOD1G93A ALS transgenic mice) in vitro. In primary motor neurons, NSC-34 cells and astrocytes, MSC conditioned medium (MSC CM) attenuated staurosporine (STS) - induced apoptosis in a concentration-dependent manner. Studying MSC CM-induced expression of neurotrophic factors in astrocytes and NSC-34 cells, we found that glial cell line-derived neurotrophic factor (GDNF) and ciliary neurotrophic factor (CNTF) gene expression in astrocytes were significantly enhanced by MSC CM, with differential responses of non-transgenic and mutant astrocytes. Expression of Vascular Endothelial Growth Factor (VEGF) in NSC-34 cells was significantly upregulated upon MSC CM-treatment. MSC CM significantly reduced the expression of the cytokines TNFα and IL-6 and iNOS both in transgenic and non-transgenic astrocytes. Gene expression of the neuroprotective chemokine Fractalkine (CX3CL1) was also upregulated in mutant SOD1G93A transgenic astrocytes by MSC CM treatment. Correspondingly, MSC CM increased the respective receptor, CX3CR1, in mutant SOD1G93A transgenic microglia. Our data demonstrate that MSC modulate motor neuronal and glial response to apoptosis and inflammation. MSC therefore represent an interesting candidate for further preclinical and clinical evaluation in ALS.


Assuntos
Esclerose Amiotrófica Lateral/metabolismo , Astrócitos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Microglia/metabolismo , Neurônios Motores/metabolismo , Esclerose Amiotrófica Lateral/genética , Animais , Apoptose/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Células Cultivadas , Quimiocina CX3CL1/metabolismo , Fator Neurotrófico Ciliar/metabolismo , Meios de Cultivo Condicionados/farmacologia , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/metabolismo , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Microglia/efeitos dos fármacos , Neurônios Motores/efeitos dos fármacos , Estaurosporina/farmacologia
18.
Oral Oncol ; 49(12): 1103-12, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24070603

RESUMO

OBJECTIVES: Perineural invasion is a prominent clinical feature of various cancers, which causes difficulty in curative resection. Glial cell-derived neurotrophic factor (GDNF), a potent neurotrophic factor, plays an important role in the invasive and metastatic behavior of various cancers. The aim of this study was to examine the role of GDNF on oral squamous cell carcinoma. MATERIALS AND METHODS: GDNF expression in tissue samples was analyzed by immunohistochemistry. Transwell assay, zymography, Western blot, reverse transcription-PCR, and electrophoretic mobility shift assay (EMSA) were carried out to assess the effects of GDNF on oral cancer cells. RESULTS: Human oral cancer tissues showed higher GDNF expression than that in normal tissues. We also found that application of human GDNF enhanced the cell migration ability of human oral cancers. Moreover, treatment with GDNF increased matrix metalloproteinase (MMP)-9 and MMP-13 expression in oral cancer. Inhibition of MMP-9 and MMP-13 in oral cancer cells by pharmacological inhibitors or neutralizing antibodies reduced GDNF-enhanced cell migration. Moreover, transfection with siRNA against MMP-13 inhibited GDNF-enhanced cell migration. Treatment with GDNF also increased ERK, p38 and JNK phosphorylation, and AP-1 DNA binding activity in human oral cancer cells. Inhibition of MAP kinase or AP-1 also reduced GDNF-induced oral cancer cell migration. In migration-prone sublines, oral cancer cells showed a higher migration ability than that of the original oral cancer cells. Surprisingly, the enhancement of cell migratory activity in migration-prone sublines was reduced by a GDNF-neutralizing antibody. Importantly, migration-prone sublines of oral cancer revealed higher GDNF expression. CONCLUSION: These results indicate a regulatory effect on cell migration by GDNF in oral squamous cancer.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Movimento Celular/fisiologia , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/metabolismo , Neoplasias Bucais/metabolismo , Anticorpos Neutralizantes/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/efeitos dos fármacos , Humanos , Metaloproteinase 13 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz/farmacologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Fator de Transcrição AP-1/antagonistas & inibidores , Fator de Transcrição AP-1/metabolismo
19.
Yi Chuan ; 35(4): 495-501, 2013 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-23659940

RESUMO

Recently, it is unclear about the mechanism of notable regenerated ability of adult zebrafish after spinal cord injury. To investigate the effects of brain on restoration from spinal cord injury, adult zebrafish spinal cord injury model was built and brain samples were dissected at different time points after the injury. Real-time quantitative PCR and in situ hybridization were applied to reveal the dynamics of glial cell line-derived neurotrophic factor (gdnf) and nitric oxide synthases (nos) mRNA expression in various regions of zebrafish brain. The results showed that, compared to sham group at each time points separately, the expression of gdnf mRNA in adult zebrafish brain during both acute phase (4 h and 12 h) and chronic phase of neuroregeneration (6 d and 11 d) increased significantly (P<0.05). The expression of nos mRNA in zebrafish brain enhanced during acute phase, and then reduced to the level lower than the sham group during the chronic phase of neuroregeneration (11 d) (P<0.05). This suggests that brain may promote neural axons regeneration in spinal cord via a more beneficial microenvironment which retains higher level of gdnf and lower level of nos.


Assuntos
Encéfalo/metabolismo , Regulação Enzimológica da Expressão Gênica , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/genética , Óxido Nítrico Sintase Tipo I/genética , Regeneração/genética , Traumatismos da Medula Espinal/fisiopatologia , Peixe-Zebra/genética , Animais , Encéfalo/citologia , Núcleo Celular/metabolismo , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/patologia , Fatores de Tempo , Regulação para Cima , Peixe-Zebra/fisiologia
20.
Cell Transplant ; 22(9): 1529-39, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23051605

RESUMO

Degeneration of midbrain dopamine neurons causes the striatal dopamine deficiency responsible for the hallmark motor symptoms of Parkinson's disease (PD). Intraparenchymal delivery of neurotrophic factors, such as glial cell line-derived neurotrophic factor (GDNF), is a possible future therapeutic approach. In animal PD models, GDNF can both ameliorate neurodegeneration and promote recovery of the dopamine system following a toxic insult. However, clinical studies have generated mixed results, and GDNF has not been efficacious in genetic animal models based on α-synuclein overexpression. We have tested the response to GDNF in a genetic mouse PD model with progressive degeneration of dopamine neurons caused by mitochondrial impairment. We find that GDNF, delivered to the striatum by either an adeno-associated virus or via miniosmotic pumps, partially alleviates the progressive motor symptoms without modifying the rate of neurodegeneration. These behavioral changes are accompanied by increased levels of dopamine in the midbrain, but not in striatum. At high levels, GDNF may instead reduce striatal dopamine levels. These results demonstrate the therapeutic potential of GDNF in a progressively impaired dopamine system.


Assuntos
Terapia Genética/métodos , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/administração & dosagem , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/genética , Doença de Parkinson/terapia , Adenoviridae/genética , Animais , Modelos Animais de Doenças , Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/biossíntese , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Imuno-Histoquímica , Masculino , Camundongos , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Distribuição Aleatória
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...