Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Intervalo de año de publicación
1.
Crit Rev Eukaryot Gene Expr ; 34(6): 71-78, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38912964

RESUMEN

Esophageal squamous cell carcinoma (ESCC) is a common malignancy of the gastrointestinal tract with a single therapeutic option and a lack of effective clinical therapeutic biomarkers. Extracellular matrix (ECM) remodeling plays a pro-carcinogenic role in a variety of malignancies, but its role in esophageal squamous carcinoma remains to be elucidated. In this study, we examined the expression levels of ECM remodeling markers in 71 pairs of esophageal squamous carcinoma tissues and normal tissues adjacent to the carcinoma using immunohistochemical staining, and analyzed their relationship with clinicopathological features and prognosis. The results suggested that extracellular matrix remodeling markers (integrin αV, fibronectin, MMP9) were abnormally highly expressed in esophageal squamous carcinoma tissues. There was a statistically significant difference between the positive expression of ECM remodeling and the TNM stage of esophageal squamous carcinoma, and there was no statistically significant correlation with age, gender and carcinoembryonic antigen expression, differentiation degree, T stage, and lymph node metastasis. Overall survival rate and overall survival time were significantly lower in patients with positive ECM remodeling expression, which was an independent risk factor for poor prognosisof esophageal squamous carcinoma.


Asunto(s)
Biomarcadores de Tumor , Carcinoma de Células Escamosas , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Matriz Extracelular , Fibronectinas , Humanos , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/genética , Masculino , Femenino , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/patología , Carcinoma de Células Escamosas de Esófago/genética , Matriz Extracelular/metabolismo , Pronóstico , Persona de Mediana Edad , Fibronectinas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/genética , Anciano , Metaloproteinasa 9 de la Matriz/metabolismo , Integrina alfaV/metabolismo , Integrina alfaV/genética , Estadificación de Neoplasias , Regulación Neoplásica de la Expresión Génica , Metástasis Linfática , Adulto
2.
Clin. transl. oncol. (Print) ; 25(8): 2487-2498, aug. 2023. ilus, graf
Artículo en Inglés | IBECS | ID: ibc-222425

RESUMEN

Objective Esophageal squamous cell carcinoma (ESCC) is a common and aggressive malignancy of the gastrointestinal tract for which therapeutic options are scarce. This study screens for LOXL2, a key gene in ESCC, and explains the molecular mechanism by which it promotes the progression of ESCC. Methods Immunohistochemical staining was performed to detect the expression level of LOXL2 in ESCC tissues and paraneoplastic tissues. CCK-8 and Transwell assays were performed to assess the effects of LOXL2 knockdown and overexpression on the proliferation, apoptosis, migration and invasion ability of ESCC cells. High-throughput sequencing analysis screens for molecular mechanisms of action by which LOXL2 promotes ESCC progression. Western blotting and qRT-PCR were used to determine the expression levels of relevant markers. Results LOXL2 is positively expressed in ESCC and highly correlated with poor prognosis. Silencing LOXL2 significantly inhibited the proliferation, migration and invasive ability of ESCC cells, whereas overexpression showed the opposite phenotype. High-throughput sequencing suggested that LOXL2-associated differentially expressed genes were highly enriched in the PI3K/AKT signaling pathway. In vitro cellular assays confirmed that silencing LOXL2 significantly reduced PI3K, p-AKTThr308 and p-AKTSer473 gene and protein expression levels, while overexpression increased all three gene and protein levels, while AKT gene and protein expression levels were not significantly different. Conclusion This study found that LOXL2 may regulate the PI3K/AKT signaling pathway and exert protumor effects on ESCC cells through phosphorylation of AKT. LOXL2 may be a key clinical warning biomarker or therapeutic target for ESCC (AU)


Asunto(s)
Humanos , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/patología , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Regulación Neoplásica de la Expresión Génica , Fosfatidilinositol 3-Quinasas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Proteína-Lisina 6-Oxidasa/genética , Proteína-Lisina 6-Oxidasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/genética , Fosforilación
3.
Clin Transl Oncol ; 25(8): 2487-2498, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36995521

RESUMEN

OBJECTIVE: Esophageal squamous cell carcinoma (ESCC) is a common and aggressive malignancy of the gastrointestinal tract for which therapeutic options are scarce. This study screens for LOXL2, a key gene in ESCC, and explains the molecular mechanism by which it promotes the progression of ESCC. METHODS: Immunohistochemical staining was performed to detect the expression level of LOXL2 in ESCC tissues and paraneoplastic tissues. CCK-8 and Transwell assays were performed to assess the effects of LOXL2 knockdown and overexpression on the proliferation, apoptosis, migration and invasion ability of ESCC cells. High-throughput sequencing analysis screens for molecular mechanisms of action by which LOXL2 promotes ESCC progression. Western blotting and qRT-PCR were used to determine the expression levels of relevant markers. RESULTS: LOXL2 is positively expressed in ESCC and highly correlated with poor prognosis. Silencing LOXL2 significantly inhibited the proliferation, migration and invasive ability of ESCC cells, whereas overexpression showed the opposite phenotype. High-throughput sequencing suggested that LOXL2-associated differentially expressed genes were highly enriched in the PI3K/AKT signaling pathway. In vitro cellular assays confirmed that silencing LOXL2 significantly reduced PI3K, p-AKTThr308 and p-AKTSer473 gene and protein expression levels, while overexpression increased all three gene and protein levels, while AKT gene and protein expression levels were not significantly different. CONCLUSION: This study found that LOXL2 may regulate the PI3K/AKT signaling pathway and exert protumor effects on ESCC cells through phosphorylation of AKT. LOXL2 may be a key clinical warning biomarker or therapeutic target for ESCC.


Asunto(s)
Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Humanos , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias Esofágicas/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína-Lisina 6-Oxidasa/genética , Proteína-Lisina 6-Oxidasa/metabolismo , Fosforilación , Movimiento Celular , Transducción de Señal/genética , Proliferación Celular , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica
4.
Materials (Basel) ; 14(4)2021 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-33670015

RESUMEN

MgO-doped LiNbO3 (LN:Mg) is famous for its high resistance to optical damage, but this phenomenon only occurs in visible and infrared regions, and its photorefraction is not decreased but enhanced in ultraviolet region. Here we investigated a series of ZrO2 co-doped LN:Mg (LN:Mg,Zr) regarding their ultraviolet photorefractive properties. The optical damage resistance experiment indicated that the resistance against ultraviolet damage of LN:Mg was significantly enhanced with increased ZrO2 doping concentration. Moreover, first-principles calculations manifested that the enhancement of ultraviolet damage resistance for LN:Mg,Zr was mainly determined by both the increased band gap and the reduced ultraviolet photorefractive center O2-/-. So, LN:Mg,Zr crystals would become an excellent candidate for ultraviolet nonlinear optical material.

5.
Am J Transl Res ; 12(9): 5399-5415, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33042427

RESUMEN

The diagnosis of lymph node metastasis (LNM) by liquid biopsy is a novel concept prompted by the necessity to develop a more convenient and accurate method to guide the clinical management of early LNM in papillary thyroid carcinoma (PTC). However, the sensitivity and specificity of many biomarkers are not high enough. We aimed to detect circRNAs from peripheral circulation that may be better associated with the prognosis of LNM in PTC. First, Galectin-3 (Gal3) in blood was determined to be highly expressed in LNM patients. Second, based on a bioinformatics analysis and miRNA sequencing analysis from 2 paired primary and LNM tumors, miR-873 was identified to directly target Gal3, which was significantly associated with clinical parameters including LNM. Third, from additional circRNA sequencing, circRNA-UMAD1 was selected as a specific sponge for miR-873 and was correlated with Gal3 levels in peripheral circulation. Fourth, circRNA-UMAD1 and Gal3 were identified to have stronger co-biomarker potential with relatively high expression in the serum of LNM patients compared with primary tumor patients, as demonstrated by the RNA expression levels in the serum of 50 PTC patients with or without LNM by quantitative real-time PCR. Overall, the combination of circRNA-UMAD1 and Gal3 is a useful and effective co-biomarker for the prognosis of LNM in PTC patients. This new molecular typing method for LNM in PTC is more precise.

6.
Materials (Basel) ; 13(3)2020 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-32046259

RESUMEN

In this work, a Ni/graphene (Ni/G) electrode was designed and fabricated by plasma-enhanced chemical vapor deposition (PECVD) for the ultrasensitive recognition of d- and l-phenylalanine. Through a single-step PECVD process, the Ni/G electrode can achieve better hydrophilicity and larger catalytic surface area, which is beneficial for the electrochemical recognition of bio-objects. After surface modification with ß-cyclodextrin, the Ni/G electrode can distinguish d-phenylalanine from l-phenylalanine according to a 0.09 V peak shift in differential pulse voltammetry tests. Moreover, this Ni/G electrode achieved a detection limit as low as 1 nM and a wide linear range from 1 nM to 10 mM toward l-phenylalanine, with great storage stability and working stability.

7.
Cell Host Microbe ; 26(2): 240-251.e8, 2019 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-31350199

RESUMEN

Interorgan immunological communication is critical to connect the local-systemic innate immune response and orchestrate a homeostatic host defense. However, the factors and their roles in this process remain unclear. We find Drosophila IMD response in guts can sequentially trigger a systemic IMD reaction in the fat body. Sugar alcohols of the polyol pathway are essential for the spatiotemporal regulation of gut-fat body immunological communication (GFIC). IMD activation in guts causes elevated levels of sorbitol and galactitol in hemolymph. Aldose reductase (AR) in hemocytes, the rate-limiting enzyme of the polyol pathway, is necessary and sufficient for the increase of plasma sugar alcohols. Sorbitol relays GFIC by subsequent activation of Metalloprotease 2, which cleaves PGRP-LC to activate IMD response in fat bodies. Thus, this work unveils how GFIC relies on the intermediate activation of the polyol pathway in hemolymph and demonstrates that AR provides a critical metabolic checkpoint in the global inflammatory response.


Asunto(s)
Alarminas/inmunología , Drosophila/inmunología , Inmunidad Innata/fisiología , Polímeros/metabolismo , Alcoholes del Azúcar/metabolismo , Aldehído Reductasa/genética , Aldehído Reductasa/metabolismo , Aldo-Ceto Reductasas/genética , Animales , Animales Modificados Genéticamente , Proteínas Portadoras/metabolismo , Drosophila/genética , Cuerpo Adiposo/metabolismo , Galactitol/sangre , Galactitol/metabolismo , Hemolinfa/metabolismo , Humanos , Inflamación/inmunología , Masculino , Metaloproteasas/metabolismo , Transducción de Señal/inmunología , Sorbitol/sangre , Sorbitol/metabolismo , Alcoholes del Azúcar/sangre
8.
J Vis Exp ; (145)2019 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-30933079

RESUMEN

Virus spreading is a major cause of epidemic diseases. Thus, understanding the interaction between the virus and the host is very important to extend our knowledge of prevention and treatment of viral infection. The fruit fly Drosophila melanogaster has proven to be one of the most efficient and productive model organisms to screen for antiviral factors and investigate virus-host interaction, due to powerful genetic tools and highly conserved innate immune signaling pathways. The procedure described here demonstrates a nano-injection method to establish viral infection and induce systemic antiviral responses in adult flies. The precise control of the viral injection dose in this method enables high experimental reproducibility. Protocols described in this study include the preparation of flies and the virus, the injection method, survival rate analysis, the virus load measurement, and an antiviral pathway assessment. The influence effects of viral infection by the flies' background were mentioned here. This infection method is easy to perform and quantitatively repeatable; it can be applied to screen for host/viral factors involved in virus-host interaction and to dissect the crosstalk between innate immune signaling and other biological pathways in response to viral infection.


Asunto(s)
Drosophila melanogaster/virología , Interacciones Huésped-Patógeno , Virosis/virología , Animales , Antivirales/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Inmunidad Innata/inmunología , Mutación/genética , Interferencia de ARN , Reproducibilidad de los Resultados , Transducción de Señal , Carga Viral , Virosis/inmunología , Virus/metabolismo , Wolbachia/fisiología
9.
J Virol ; 92(18)2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29976667

RESUMEN

In order to establish productive infection and dissemination, viruses usually evolve a number of strategies to hijack and/or subvert the host defense systems. However, host factors utilized by the virus to facilitate infection remain poorly characterized. In this work, we found that Drosophila melanogaster deficient in budding uninhibited by benzimidazoles 1 (bub1), a highly conserved subunit of the kinetochore complex regulating chromosome congression (1), became resistant to Drosophila C virus (DCV) infection, evidenced in increased survival rates and reduced viral loads, compared to the wild-type control. Mechanistic analysis further showed that Bub1 also functioned in the cytoplasm and was essentially involved in clathrin-dependent endocytosis of DCV and other pathogens, thus limiting pathogen entry. DCV infection potentially had strengthened the interaction between Bub1 and the clathrin adaptor on the cell membrane. Furthermore, the conserved function of Bub1 was also verified in a mammalian cell line. Thus, our data demonstrated a previously unknown function of Bub1 that could be hijacked by pathogens to facilitate their infection and spread.IMPORTANCE In this work, we identify for the first time that the nuclear protein Bub1 (budding uninhibited by benzimidazoles 1), a highly conserved subunit of the kinetochore complex regulating chromosome congression, has a novel and important function on the cell membrane to facilitate the virus to enter host cells. Bub1 deficiency empowers the host to have the ability to resist viral infection in Drosophila and a human cell line. Bub1 is involved in the virus entry step through regulating endocytosis. The DCV capsid protein can recruit Bub1, and DCV infection can strengthen the interaction between Bub1 and a clathrin-dependent endocytosis component. The restricted entry of vesicular stomatitis virus (VSV) and Listeria monocytogenes in bub1-deficient flies and cell lines was also observed. Therefore, our data implicate a previously unknown function of Bub1 that can be hijacked by pathogens to facilitate their entry, and Bub1 may serve as a potential antiviral therapy target for limiting viral entry.


Asunto(s)
Dicistroviridae/patogenicidad , Drosophila melanogaster/virología , Endocitosis , Proteínas Serina-Treonina Quinasas/genética , Internalización del Virus , Regiones no Traducidas 3'/fisiología , Animales , Clatrina/metabolismo , Dicistroviridae/genética , Dicistroviridae/fisiología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Células HEK293 , Interacciones Huésped-Patógeno/genética , Humanos , Listeria monocytogenes/patogenicidad , Listeria monocytogenes/fisiología , Proteínas Serina-Treonina Quinasas/deficiencia , Virus de la Estomatitis Vesicular Indiana/fisiología , Carga Viral
10.
Exp Cell Res ; 368(2): 248-257, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29750899

RESUMEN

The persistent proliferation of hypoxia-induced vascular smooth muscle cells (VSMCs) in the arterial wall underlie the development of atherosclerosis. However, the mechanism that regulates the behavior of VSMCs, which involve in actin aggregation, and impedes their migration is still elusive. Here, we report that bone morphogenetic protein 2 (BMP-2) leads to enrichment of CD44 and F-actin stress fiber and secretion of matrix metalloproteinases-2 (MMP-2) during hypoxia in vitro and following artificial hypoxia-induced atherosclerosis exacerbation in vivo. To test this hypothesis, fluorescence immunostaining, immune-hybridization and flow cytometry analyses were performed to understand the relationship among BMP-2, CD44 and MMP-2 linkage. The cellular actin cytoskeleton was reduced, and smaller adhesion plaques were formed in hypoxia-induced T/G HA-VSMC cell line, but BMP-2 against disruption of F-actin and increase the motility and migration behaviors of VSMC during hypoxic cultured. Aggregation of F-actin dependents on the interaction between the cell surface integral membrane protein CD44 and Vinculin which enhanced by rBMP-2. This activity of Actin/CD44/ linkage was inhibited by competing with the active site of the CD44 using recombined the hemopexin-like C-terminal domain (PEX) of MMP-2. These results lead to the proliferation and migration of VSMCs were inhibited in response to MMP-2 activity when the cell is in a hypoxic environment. Collectively, our discovery indicates that BMP-2 could enhance migration and proliferation of hypoxia-induced VSMCs via the Actin/CD44/MMP-2 molecular pathway.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteína Morfogenética Ósea 2/metabolismo , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Receptores de Hialuranos/metabolismo , Hipoxia/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Animales , Membrana Celular/metabolismo , Células Cultivadas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Ratas , Ratas Sprague-Dawley
11.
Sci Rep ; 7(1): 5166, 2017 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-28701769

RESUMEN

Perfluorooctane sulfonate (PFOS) is an environmental contaminant that has been manufactured to be used as surfactants and repellents in industry. Due to long half-life for clearance and degradation, PFOS is accumulative in human body and has potential threat to human health. Previous studies have shown the development and function of immune cells can be affected by PFOS. Although PFOS has a high chance of being absorbed through the oral route, whether and how PFOS affects immune cells in the gut is unknown. Using mouse model of Citrobacter rodentium infection, we investigated the role of PFOS on intestinal immunity. We found at early phase of the infection, PFOS inhibited the expansion of the pathogen by promoting IL-22 production from the group 3 innate lymphoid cell (ILC3) in an aryl hydrocarbon receptor dependent manner. Nevertheless, persistent PFOS treatment in mice finally led to a failure to clear the pathogen completely. At late phase of infection, enhanced bacterial counts in PFOS treated mice were accompanied by increased inflammatory cytokines, reduced mucin production and dysbiosis, featured by decreased level of Lactobacillus casei, Lactobacillus johnsonii and increased E. coli. Our study reveals a deleterious consequence in intestinal bacterial infection caused by PFOS accumulation.


Asunto(s)
Ácidos Alcanesulfónicos/farmacología , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/microbiología , Fluorocarburos/farmacología , Intestinos/efectos de los fármacos , Intestinos/inmunología , Intestinos/microbiología , Animales , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Ratones , Mucinas/metabolismo , Índice de Severidad de la Enfermedad , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th17/metabolismo
12.
Sci Rep ; 5: 7961, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-25609658

RESUMEN

PACT and its murine ortholog RAX were originally identified as a protein activator for the dsRNA-dependent, interferon-inducible protein kinase PKR. Recent studies indicated that RAX played a role in embryogenesis and neuronal development. In this study, we investigated the expression of RAX during the postnatal development of the mouse cerebellum and its role in the migration of cerebellar granule neurons (CGNs). High expression of RAX was observed in the cerebellum from postnatal day (PD) 4 to PD9, a period when the CGNs migrate from the external granule layer (EGL) to the internal granule layer (IGL). The migration of the EGL progenitor cells in vivo was inhibited by RAX knockdown on PD4. This finding was confirmed by in vitro studies showing that RAX knockdown impaired the migration of CGNs in cerebellar microexplants. PACT/RAX-regulated migration required its third motif and was independent of PKR. PACT/RAX interacted with focal adhesion kinase (FAK) and PACT/RAX knockdown disturbed the FAK phosphorylation in CGNs. These findings demonstrated a novel function of PACT/RAX in the regulation of neuronal migration.


Asunto(s)
Proteínas Portadoras/metabolismo , Movimiento Celular , Cerebelo/crecimiento & desarrollo , Gránulos Citoplasmáticos/metabolismo , Proteínas del Ojo/metabolismo , Proteínas de Homeodominio/metabolismo , Neuronas/citología , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Secuencias de Aminoácidos , Animales , Diferenciación Celular , Proliferación Celular , Proteínas del Ojo/química , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/química , Ratones Endogámicos C57BL , Neuroglía/citología , Neuroglía/metabolismo , Fosforilación , Unión Proteica , Factores de Transcripción/química , eIF-2 Quinasa/metabolismo
13.
J Immunol ; 193(5): 2157-67, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25063874

RESUMEN

Multiple sclerosis (MS) is a complex multifactorial disease that results from the interplay between environmental factors and a susceptible genetic background. Experimental autoimmune encephalomyelitis (EAE) has been widely used to investigate the mechanisms underlying MS pathogenesis. Chemokines, such as CCL2, are involved in the development of EAE. We have previously shown that thiamine deficiency (TD) induced CCL2 in neurons. We hypothesized that TD may affect the pathogenesis of EAE. In this study, EAE was induced in C57BL/6J mice by the injection of myelin oligodendroglial glycoprotein (MOG) peptides 35-55 with or without TD. TD aggravated the development of EAE, which was indicated by clinical scores and pathologic alterations in the spinal cord. TD also accelerated the development of EAE in an adoptive transfer EAE model. TD caused microglial activation and a drastic increase (up 140%) in leukocyte infiltration in the spinal cord of the EAE mice; specifically, TD increased Th1 and Th17 cells. TD upregulated the expression of CCL2 and its receptor CCR2 in the spinal cord of EAE mice. Cells in peripheral lymph node and spleen isolated from MOG-primed TD mice showed much stronger proliferative responses to MOG. CCL2 stimulated the proliferation and migration of T lymphocytes in vitro. Our results suggested that TD exacerbated the development of EAE through activating CCL2 and inducing pathologic inflammation.


Asunto(s)
Movimiento Celular/inmunología , Quimiocina CCL2/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Células TH1/inmunología , Células Th17/inmunología , Deficiencia de Tiamina/inmunología , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Encefalomielitis Autoinmune Experimental/complicaciones , Encefalomielitis Autoinmune Experimental/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ratones , Microglía/inmunología , Microglía/patología , Glicoproteína Mielina-Oligodendrócito/inmunología , Glicoproteína Mielina-Oligodendrócito/toxicidad , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , Ratas , Médula Espinal/inmunología , Médula Espinal/patología , Bazo/inmunología , Bazo/patología , Células TH1/patología , Células Th17/patología , Deficiencia de Tiamina/complicaciones , Deficiencia de Tiamina/patología
14.
Mol Neurodegener ; 8: 27, 2013 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-23938027

RESUMEN

BACKGROUND: Accumulation of ß-amyloid peptides is an important hallmark of Alzheimer's disease (AD). Tremendous efforts have been directed to elucidate the mechanisms of ß-amyloid peptides degradation and develop strategies to remove ß-amyloid accumulation. In this study, we demonstrated that a subpopulation of oligodendroglial precursor cells, also called NG2 cells, were a new cell type that can clear ß-amyloid peptides in the AD transgene mice and in NG2 cell line. RESULTS: NG2 cells were recruited and clustered around the amyloid plaque in the APPswe/PS1dE9 mice, which is Alzheimer's disease mouse model. In vitro, NG2 cell line and primary NG2 cells engulfed ß-amyloid peptides through the mechanisms of endocytosis in a time dependent manner. Endocytosis is divided into pinocytosis and phagocytosis. Aß(42) internalization by NG2 cells was mediated by actin-dependent macropinocytosis. The presence of ß-amyloid peptides stimulated the autophagic pathway in NG2 cells. Once inside the cells, the ß-amyloid peptides in NG2 cells were transported to lysosomes and degraded by autophagy. CONCLUSIONS: Our findings suggest that NG2 cells are a new cell type that can clear ß-amyloid peptides through endocytosis and autophagy.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Autofagia/fisiología , Células-Madre Neurales/metabolismo , Oligodendroglía/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Citometría de Flujo , Humanos , Immunoblotting , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Placa Amiloide/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa
15.
J Neurochem ; 126(6): 805-18, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23586593

RESUMEN

Thiamine deficiency (TD) causes mild impairment of oxidative metabolism and region-selective neuronal loss in the brain, which may be mediated by neuronal oxidative stress, endoplasmic reticulum (ER) stress, and neuroinflammation. TD-induced brain damage is used to model neurodegenerative disorders, and the mechanism for the neuronal death is still unclear. We hypothesized that autophagy might be activated in the TD brain and play a protective role in TD-induced neuronal death. Our results demonstrated that TD induced the accumulation of autophagosomes in thalamic neurons measured by transmission electron microscopy, and the up-regulation of autophagic markers LC3-II, Atg5, and Beclin1 as measured with western blotting. TD also increased the expression of autophagic markers and induced LC3 puncta in SH-SY5Y neuroblastoma cells. TD-induced expression of autophagic markers was reversed once thiamine was re-administered. Both inhibition of autophagy by wortmannin and Beclin1 siRNA potentiated TD-induced death of SH-SY5Y cells. In contrast, activation of autophagy by rapamycin alleviated cell death induced by TD. Intraperitoneal injection of rapamycin stimulated neuronal autophagy and attenuated TD-induced neuronal death and microglia activation in the submedial thalamus nucleus (SmTN). TD inhibited the phosphorylation of p70S6 kinase, suggesting mTOR/p70S6 kinase pathway was involved in the TD-induced autophagy. These results suggest that autophagy is neuroprotective in response to TD-induced neuronal death in the central nervous system. This opens a potential therapeutic avenue for neurodegenerative diseases caused by mild impairment of oxidative metabolism. Autophagy is neuroprotective in response to thiamine deficiency (TD)-induced neuronal death. TD caused neuronal damage and induced the formation of autophagosome, and increased the expression of autophagy-related proteins. Autophagy sequestered damaged and dysfunctional organelles/protein, and transported them to lysosomes for degradation/recycling. This process provided nutrients for injured neurons. Wortmannin and knockdown of Beclin1 inhibited autophagy, and exacerbated TD-induced cell death, while activation of autophagy by rapamycin offered protection against TD neurotoxicity.


Asunto(s)
Autofagia/fisiología , Degeneración Nerviosa/patología , Androstadienos/farmacología , Animales , Antibacterianos/toxicidad , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/farmacología , Beclina-1 , Western Blotting , Muerte Celular/efectos de los fármacos , Línea Celular , Regulación hacia Abajo , Humanos , Inmunohistoquímica , Masculino , Proteínas de la Membrana/farmacología , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Proteínas Asociadas a Microtúbulos/metabolismo , Oxidación-Reducción , Fagosomas/metabolismo , ARN Interferente Pequeño/genética , Sirolimus/toxicidad , Tálamo/citología , Tálamo/metabolismo , Transfección , Vacuolas/metabolismo , Wortmanina
16.
J Neurosci Res ; 89(10): 1676-84, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21671257

RESUMEN

Ethanol exposure induces neurodegeneration in the developing central nervous system (CNS). Fetal alcohol spectrum disorders (FASD) are caused by ethanol exposure during pregnancy and are the most common nonhereditary cause of mental retardation. It is important to identify agents that provide neuroprotection against ethanol neurotoxicity. Multiple mechanisms have been proposed for ethanol-induced neurodegeneration, and oxidative stress is one of the most important mechanisms. Recent evidence indicates that glycogen synthase kinase 3ß (GSK3ß) is a potential mediator of ethanol-mediated neuronal death. Cyanidin-3-glucoside (C3G), a member of the anthocyanin family, is a potent natural antioxidant. Our previous study suggested that C3G inhibited GSK3ß activity in neurons. Using a third trimester equivalent mouse model of ethanol exposure, we tested the hypothesis that C3G can ameliorate ethanol-induced neuronal death in the developing brain. Intraperitoneal injection of C3G reduced ethanol-meditated caspase-3 activation, neurodegeneration, and microglial activation in the cerebral cortex of 7-day-old mice. C3G blocked ethanol-mediated GSK3ß activation by inducing phosphorylation at serine 9 while reducing the phosphorylation at tyrosine 216. C3G also inhibited ethanol-stimulated expression of malondialdehyde (MDA) and p47phox, indicating that C3G alleviated ethanol-induced oxidative stress. These results provide important insight into the therapeutic potential of C3G.


Asunto(s)
Trastornos del Sistema Nervioso Inducidos por Alcohol/tratamiento farmacológico , Antocianinas/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/embriología , Etanol/antagonistas & inhibidores , Trastornos del Espectro Alcohólico Fetal/tratamiento farmacológico , Glucósidos/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Modelos Animales de Enfermedad , Etanol/toxicidad , Femenino , Ratones , Ratones Endogámicos C57BL , Embarazo
17.
Alcohol Clin Exp Res ; 35(9): 1574-83, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21599712

RESUMEN

BACKGROUND: Ethanol exposure during brain development causes profound damages to the central nervous system (CNS). The underlying cellular/molecular mechanisms remain unclear. The endoplasmic reticulum (ER) is involved in posttranslational protein processing and transport. The accumulation of unfolded or misfolded proteins in the ER lumen triggers ER stress, which is characterized by translational attenuation, synthesis of ER chaperone proteins, and activation of transcription factors. Sustained ER stress ultimately leads to cell death. ER stress is implicated in various neurodegenerative processes. METHODS: Using a third trimester equivalent mouse model of ethanol exposure, we tested the hypothesis that ethanol induces ER stress in the developing brain. Seven-day-old C57BL/6 mice were acutely exposed to ethanol by subcutaneous injection and the expression of ER stress-inducible proteins (ERSIPs) and signaling pathways associated with ER stress were examined. RESULTS: Ethanol exposure significantly increased the expression of ERSIPs and activated signaling pathways associated with ER stress; these include ATF6, CHOP/GADD153, GRP78, and mesencephalic astrocyte-derived neurotrophic factor as well as the phosphorylation of IRE1α, eIF2α, PERK, and PKR. The ethanol-induced increase in ERSIPs occurred within 4 hours of ethanol injection, and levels of some ERSIPs remained elevated after 24 hours of ethanol exposure. Ethanol-induced increase in phosphorylated eIF2α, caspase-12, and CHOP was distributed in neurons of specific areas of the cerebral cortex, hippocampus, and thalamus. CONCLUSIONS: Our finding indicates that ethanol induces ER stress in immature neurons, providing novel insight into ethanol's detrimental effect on the developing CNS.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Corteza Cerebral/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Retículo Endoplásmico/metabolismo , Etanol/farmacología , Actinas/metabolismo , Animales , Caspasa 3/biosíntesis , Depresores del Sistema Nervioso Central/sangre , Depresores del Sistema Nervioso Central/metabolismo , Chaperón BiP del Retículo Endoplásmico , Etanol/sangre , Etanol/metabolismo , Proteínas de Choque Térmico/biosíntesis , Hipocampo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Tálamo/metabolismo , Factores de Tiempo , Factor de Transcripción CHOP/biosíntesis
18.
Neurochem Res ; 36(3): 367-74, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21203835

RESUMEN

Lithium has been used for the treatment of bipolar mood disorder and is shown to have neuroprotective properties. Since lithium inhibits the activity of glycogen synthase kinase 3 (GSK3) which is implicated in various human diseases, particularly neurodegenerative diseases, the therapeutic potential of lithium receives great attention. Parkinson's disease (PD) is the second most common neurodegenerative disease, characterized by the pathological loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). Intranigral injection of the catecholaminergic neurotoxin 6-hydroxydopamine (6-OHDA) causes selective and progressive degeneration of dopaminergic neurons in SNpc, and is a commonly used animal model of PD. The current study was designated to determine whether lithium is effective in alleviating 6-OHDA-induced neurodegeneration in the SNpc of rats. We demonstrated that chronic subcutaneous administration of lithium inhibited GSK3 activity in the SNpc, which was evident by an increase in phosphorylation of GSK3ß at serine 9, cyclin D1 expression, and a decrease in tau phosphorylation. 6-OHDA did not affect GSK3 activity in the SNpc. Moreover, lithium was unable to alleviate 6-OHDA-induced degeneration of SNpc dopaminergic neurons. The results suggest that GSK3 is minimally involved in the neurodegeneration in the rat 6-OHDA model of PD.


Asunto(s)
Dopamina/metabolismo , Litio/farmacología , Neuronas/efectos de los fármacos , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Oxidopamina/farmacología , Enfermedad de Parkinson/patología , Animales , Antipsicóticos/farmacología , Antipsicóticos/uso terapéutico , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Litio/uso terapéutico , Masculino , Neuronas/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley , Simpaticolíticos/farmacología
19.
Brain Pathol ; 21(3): 279-97, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21029241

RESUMEN

Chemokines are implicated in the neuroinflammation of several chronic neurodegenerative disorders. However, the precise role of chemokines in neurodegeneration is unknown. Thiamine deficiency (TD) causes abnormal oxidative metabolism in the brain as well as a well-defined microglia activation and neurodegeneration in the submedial thalamus nucleus (SmTN), which are common features of neurodegenerative diseases. We evaluated the role of chemokines in neurodegeneration and the underlying mechanism in a TD model. Among the chemokines examined, TD selectively induced neuronal expression of monocyte chemoattractant protein-1 (MCP-1) in the SmTN prior to microglia activation and neurodegeneration. The conditioned medium collected from TD-induced neurons caused microglia activation. With a neuron/microglia co-culture system, we showed that MCP-1-induced neurotoxicity required the presence of microglia, and exogenous MCP-1 was able to activate microglia and stimulated microglia to produce cytokines. A MCP-1 neutralizing antibody inhibited MCP-1-induced microglia activation and neuronal death in culture and in the thalamus. MCP-1 knockout mice were resistant to TD-induced neuronal death in SmTN. TD selectively induced the accumulation of reactive oxygen species in neurons, and antioxidants blocked TD-induced MCP-1 expression. Together, our results indicated an induction of neuronal MCP-1 during mild impairment of oxidative metabolism caused by microglia recruitment/activation, which exacerbated neurodegeneration.


Asunto(s)
Quimiocina CCL2/metabolismo , Microglía/fisiología , Degeneración Nerviosa/metabolismo , Neuronas/metabolismo , Núcleos Talámicos Posteriores/metabolismo , Deficiencia de Tiamina/metabolismo , Animales , Muerte Celular/fisiología , Quimiocina CCL2/genética , Quimiocinas/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/inmunología , Degeneración Nerviosa/patología , Neuronas/patología , Oxidación-Reducción , Núcleos Talámicos Posteriores/citología , Núcleos Talámicos Posteriores/patología , Especies Reactivas de Oxígeno/metabolismo , Deficiencia de Tiamina/inmunología
20.
Neurobiol Aging ; 32(1): 42-53, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19233513

RESUMEN

Thiamine pyrophosphate (TPP) and the activities of thiamine-dependent enzymes are reduced in Alzheimer's disease (AD) patients. In this study, we analyzed the relationship between thiamine deficiency (TD) and amyloid precursor protein (APP) processing in both cellular and animal models of TD. In SH-SY5Y neuroblastoma cells overexpressing APP, TD promoted maturation of ß-site APP cleaving enzyme 1 (BACE1) and increased ß-secretase activity which resulted in elevated levels of ß-amyloid (Aß) as well as ß-secretase cleaved C-terminal fragment (ß-CTF). An inhibitor of ß-secretase efficiently reduced TD-induced up-regulation of Aß and ß-CTF. Importantly, thiamine supplementation reversed the TD-induced alterations. Furthermore, TD treatment caused a significant accumulation of reactive oxygen species (ROS); antioxidants suppressed ROS production and maturation of BACE1, as well as TD-induced Aß accumulation. On the other hand, exogenous Aß(1-40) enhanced TD-induced production of ROS. A study on mice indicated that TD also caused Aß accumulation in the brain, which was reversed by thiamine supplementation. Taken together, our study suggests that TD could enhance Aß generation by promoting ß-secretase activity, and the accumulation of Aß subsequently exacerbated TD-induced oxidative stress.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Deficiencia de Tiamina/metabolismo , Deficiencia de Tiamina/patología , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/metabolismo , Análisis de Varianza , Animales , Antioxidantes/uso terapéutico , Ácido Aspártico Endopeptidasas/metabolismo , Muerte Celular/fisiología , Línea Celular Tumoral , Cromanos/uso terapéutico , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuroblastoma/patología , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Piritiamina/efectos adversos , Especies Reactivas de Oxígeno/metabolismo , Tiamina/administración & dosificación , Deficiencia de Tiamina/tratamiento farmacológico , Deficiencia de Tiamina/etiología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...