Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Inflamm Regen ; 43(1): 49, 2023 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-37833789

RESUMEN

BACKGROUND: Testosterone contributes to male organism development, such as bone density, muscle development, and fat repartition. Estrogen (derived from testosterone) also contributes to female reproductive system development. Here, we investigated the effect of testosterone on glioma cells and brain neuron inflammation essential for cancer development and progression. METHODS: The human astrocyte and glioma cell lines were treated with 6 ng/ml exogenous testosterone in vitro. We performed cell counting kit-8, transwell, and wound healing assays to determine the effect of testosterone on glioma cell proliferation, migration, and invasion. The glioma cells were injected into the xenograft and treated with 5 µl concentrated testosterone. Transcriptional suppression of glial cell line-derived neurotrophic factor (GDNF) was performed to evaluate brain neuron inflammation and survival. The tumor tissues were assessed by hematoxylin-eosin staining and immunohistochemistry. RESULTS: Testosterone upregulates GDNF to stimulate proliferation, migration, and invasion of glioma cells. Pathologically, the augmentation of GDNF and cyclophilin A contributed to neuroprotection when treated with testosterone. Our investigation showed that testosterone contributes to brain neuron and astrocyte inflammation through the upregulation of nuclear factor erythroid 2-related factor 2 (NRF2), glial fibrillary acid protein (GFAP), and sirtuin 5 (SIRT5), resulting in pro-inflammatory macrophages recruitments into the neural microenvironment. Mechanically, testosterone treatment regulates GDNF translocation from the glioma cells and astrocyte nuclei to the cytoplasm. CONCLUSION: Testosterone upregulates GDNF in glioma cells and astrocytes essential for microglial proliferation, migration, and invasion. Testosterone contributes to brain tumor growth via GDNF and inflammation. The contribution of testosterone, macrophages, and astrocytes, in old neuron rescue, survival, and proliferation. During brain neuron inflammation, the organism activates and stimulates the neuron rescue through the enrichment of the old neuron microenvironment with growth factors such as GDNF, BDNF, SOX1/2, and MAPK secreted by the surrounding neurons and glial cells to maintain the damaged neuron by inflammation alive even if the axon is dead. The immune response also contributes to brain cell survival through the secretion of proinflammatory cytokines, resulting in inflammation maintenance. The rescued old neuron interaction with infiltrated macrophages contributes to angiogenesis to supplement the old neuron with more nutrients leading to metabolism activation and surrounding cell uncontrollable cell growth.

2.
J Cell Physiol ; 235(12): 9347-9360, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32356318

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disease characterized by progressive loss of dopaminergic neurons in the substantia nigra and striatum. Glial cell line-derived neurotrophic factor (GDNF) can effectively promote the differentiation and survival of many types of neurons, especially dopaminergic neurons, suggesting it could be a treatment for PD. Lipid rafts are highly dynamic cell membrane domains that contain numerous signal protein receptors, providing an important platform for signal transduction. Compelling evidence indicates that alterations in lipid rafts are associated with PD, and some studies have reported that GDNF can regulate the expression of caveolin-1, a lipid raft-marker protein. However, the precise effects of GDNF on lipid rafts remain unknown. We developed a cellular PD model, purified detergent-resistant membranes (membrane rafts), and performed proteomic and lipid metabolomics analyses to examine changes in lipid rafts after GDNF treatment. The results showed considerable protein and lipid alterations in response to GDNF, especially altered levels of dopamine-ß-hydroxylase, heat shock 70 kDa protein, neural cell adhesion molecule, cytoskeletal proteins, and long-chain polysaturated/unsaturated fatty acids. These findings reveal a new avenue to explore the relationships between GDNF, lipid rafts, and PD and support the hypothesis that GDNF may be a useful treatment for PD.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Lípidos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Supervivencia Celular/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Metabolismo de los Lípidos/fisiología , Microdominios de Membrana/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo
3.
Immunol Invest ; 49(1-2): 134-145, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31469350

RESUMEN

A disintegrin and metalloproteinases (ADAMs) are believed to be involved in the pathogenesis of many fibrosis-related diseases. However, little is known regarding the significance of ADAM17 as a biomarker for interstitial lung disease (ILD). In this study, by using the RT-PCR, western blotting and ELISA, we detected the expression level of ADAM17 in peripheral blood mononuclear cells and serum from idiopathic pulmonary fibrosis (IPF) patients, connective tissue disease associated ILD (CTD-ILD) patients and healthy controls, and correlations between clinical and laboratory parameters were also analyzed. We found that IPF patients and CTD-ILD patients showed higher levels of ADAM17 than healthy controls. Moreover, ADAM17 in IPF patients with acute exacerbation (AE-IPF) was significantly higher than that in stable IPF (S-IPF) patients. Expression of ADAM17 was positively correlated with disease duration and CRP but negatively correlated with diffusing capacity of carbon monoxide (DLCO) and total lung capacity (TLC). Among the CTD-ILD patients, SSc-ILD patients had the highest serum levels of ADAM17 compared with the RA-ILD, SS-ILD and IIM-ILD groups and ADAM17 expression levels were correlated with image grading. In conclusion, this study showed that ADAM17 is highly expressed in ILD patients and is associated with disease activity and severity. Additionally, ADAM17 expression is not only related to the primary CTDs, but also to image grading. ADAM17 may serve as a new biomarker for ILD.


Asunto(s)
Proteína ADAM17/biosíntesis , Biomarcadores/sangre , Enfermedades Pulmonares Intersticiales/metabolismo , Proteína ADAM17/sangre , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
4.
Oncotarget ; 8(43): 74019-74035, 2017 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-29088765

RESUMEN

The aim of this study was to identify the receptor for glial cell line-derived neurotrophic factor (GDNF) in glioblastoma multiforme (GBM). After GST pull-down assays, membrane proteins purified from C6 rat glioma cells were subjected to liquid chromatography-tandem mass spectrometry (LC-MS/MS). The differentially expressed proteins were annotated using Gene Ontology, and neuropilin-1 (NRP1) was identified as the putative GDNF receptor in glioma. NRP1 was more highly expressed in human GBM brains and C6 rat glioma cells than in normal human brains or primary rat astrocytes. Immunofluorescence staining showed that NRP1 was recruited to the membrane by GDNF, and NRP1 co-immunoprecipitated with GDNF. Using the NRP1 and GDNF protein structures to assess molecular docking in the ZDOCK server and visualization with the PyMOL Molecular Graphics System revealed 8 H-bonds and stable positive and negative electrostatic interactions between NRP1 and GDNF. RNAi knockdown of NRP1 reduced proliferation of C6 glioma cells when stimulated with GDNF. NRP1 was an independent risk factor for both survival and recurrence in GBM patients. High NRP1 mRNA expression correlated with shorter OS and DFS (OS: χ2=4.6720, P=0.0307; DFS: χ2=11.013, P=0.0009). NRP1 is thus a GDNF receptor in glioma cells and a potential therapeutic target.

5.
Int J Mol Sci ; 18(9)2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28880247

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF), a potential therapeutic factor for Parkinson's disease (PD), exerts its biological effects through the Ret receptor tyrosine kinase. The redistribution of Ret into lipid rafts substantially influences Ret signaling, but the mechanisms underlying Ret translocation remain unclear. The purpose of our study was to further explore the signaling mechanisms of GDNF and to determine whether the actin cytoskeleton is involved in the GDNF-induced Ret translocation into lipid rafts. In MN9D dopaminergic neuronal cells, we used density gradient centrifugation and immunofluorescence confocal microscopy to separate and visualize lipid rafts, co-immunoprecipitation to analyze protein-protein interactions, and latrunculin B (Lat B) and jasplakinolide (Jas) to disrupt and enhance the polymerization of the actin cytoskeleton, respectively. The results showed that Ret translocated into lipid rafts and coimmunoprecipitated with actin in response to GDNF treatment. After Lat B or Jas treatment, the Ret-F-actin association induced by GDNF was impaired or enhanced respectively and then the levels of Ret translocated into lipid rafts were correspondingly inhibited or promoted. These data indicate that actin polymerization and cytoskeletal remodeling are integral to GDNF-induced cell signaling in dopaminergic cells and define a new role of the actin cytoskeleton in promoting Ret redistribution into lipid rafts.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Proteínas Proto-Oncogénicas c-ret/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Humanos , Microdominios de Membrana , Ratones , Ratas , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA