Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cancer Invest ; 42(6): 527-537, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38965994

RESUMO

Despite the emergence of various treatment strategies for rectal cancer based on neoadjuvant chemoradiotherapy, there is currently a lack of reliable biomarkers to determine which patients will respond well to neoadjuvant chemoradiotherapy. Through collecting hematological and biochemical parameters data of patients prior to receiving neoadjuvant chemoradiotherapy, we evaluated the predictive value of systemic inflammatory indices for pathological response and prognosis in rectal cancer patients. We found that baseline GRIm-Score was an independent predictor for MPR in rectal cancer patients. However, no association was observed between several commonly systemic inflammation indices and long-term outcome.


Assuntos
Terapia Neoadjuvante , Neoplasias Retais , Humanos , Neoplasias Retais/patologia , Neoplasias Retais/terapia , Neoplasias Retais/imunologia , Masculino , Feminino , Pessoa de Meia-Idade , Terapia Neoadjuvante/métodos , Idoso , Quimioembolização Terapêutica/métodos , Prognóstico , Resultado do Tratamento , Adulto , Quimiorradioterapia/métodos
2.
Eur J Surg Oncol ; 50(4): 108242, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38460248

RESUMO

BACKGROUND: Preoperative neoadjuvant chemoradiotherapy (nCRT) followed by total mesorectal excision (TME) is a common approach for treating patients with locally advanced rectal cancer. Nevertheless, the mutational profile and its prognostic impact in surgically resected tumor specimens after nCRT remains to be clarified. METHODS: The comprehensive analysis of mutational landscape was retrospectively conducted by target regions sequencing approach that covered 150 tumor-related genes. Univariate and multivariate logistic regression and Cox regression was used to examine the association of mutation status in genes and pathways with pathological response and prognosis. Data from Memorial Sloan Kettering Cancer Center (MSK) cohort was used for comparison with our results. RESULTS: The top five commonly mutated genes in resected rectal tumor tissue samples following nCRT were TP53 (42%), APC (31%), KRAS (27%), PIK3CA (14%) and FBXW7 (11%). Mutations in the WNT pathway, which was mainly represented by APC mutation, were found to be significantly associated with tumor regression grade (TRG) 3. In our cohort, co-mutations in the receptor tyrosine kinase (RTK)/RAS and WNT pathways were found to be independently associated with reduced risk of recurrent and significantly associated with longer disease-free survival (DFS). In both our cohort and the MSK cohort, co-mutations in the TGF-ß and TP53 pathways were significantly associated with worse DFS. CONCLUSIONS: Resected rectal tumor samples from patients without complete pathological response can be appropriately used to detect mutations. Co-mutations in the TGF-ß and TP53 pathways may provide more prognostic information beyond commonly used clinical factors.


Assuntos
Terapia Neoadjuvante , Neoplasias Retais , Humanos , Prognóstico , Estudos Retrospectivos , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Quimiorradioterapia , Neoplasias Retais/genética , Neoplasias Retais/terapia , Mutação , Estadiamento de Neoplasias , Resultado do Tratamento , Proteína Supressora de Tumor p53/genética
3.
Mol Neurobiol ; 60(5): 2470-2485, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36662361

RESUMO

Triggering receptor expressed on myeloid cells-2 (TREM2), a cell surface receptor mainly expressed on microglia, has been shown to play a critical role in Alzheimer's disease (AD) pathogenesis and progression. Our recent results showed that overexpression of TREM2 inhibited inflammatory response in APP/PS1 mice and BV2 cells. Several studies indicated that TREM2 ameliorated tau hyperphosphorylation might be ascribed to the inhibition of neuroinflammation. However, the precise signaling pathways underlying the effect of TREM2 on tau pathology and neuronal apoptosis have not been fully elucidated. In the present study, upregulation of TREM2 significantly inhibited tau hyperphosphorylation at Ser199, Ser396, and Thr205, respectively, as well as prevented neuronal loss and apoptosis. We also found that upregulation of TREM2 alleviated behavioral deficits and improved the spatial cognitive ability of APP/PS1 mice. Further study revealed that TREM2 could activate phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) signaling pathway, resulting in an inhibitory effect on glycogen synthase kinase-3ß (GSK-3ß), which is a major kinase responsible for tau hyperphosphorylation in AD. In line with in vivo findings, TREM2-overexpressing BV2 microglia following ß-amyloid (Aß) stimulation led to a significant increase in the phosphorylation of PI3K, Akt, and GSK-3ß, accompanied by a decrease in tau hyperphosphorylation and apoptosis in co-cultured SH-SY5Y cells. Furthermore, LY294002, a specific PI3K inhibitor, was observed to abolish the beneficial effects of TREM2 on tau hyperphosphorylation, neuronal apoptosis, and spatial cognitive impairments in vivo and in vitro. Thus, our findings indicated that TREM2 inhibits tau hyperphosphorylation and neuronal apoptosis, at least in part, by the activation of the PI3K/Akt/GSK-3ß signaling pathway. Taken together, the above results allow us to better understand how TREM2 protects against tau pathology and suggest that upregulation of TREM2 may provide new ideas and therapeutic targets for AD.


Assuntos
Doença de Alzheimer , Neuroblastoma , Animais , Humanos , Camundongos , Doença de Alzheimer/patologia , Apoptose , Glicogênio Sintase Quinase 3 beta/metabolismo , Glicoproteínas de Membrana/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Imunológicos/metabolismo , Transdução de Sinais , Proteínas tau/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo
4.
Eur J Pharmacol ; 918: 174772, 2022 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-35090935

RESUMO

Our previous findings indicated that tanshinone IIA (tan IIA), a natural component extracted from the root and rhizome of danshen, significantly attenuated ß-amyloid accumulation, neuroinflammation, and endoplasmic reticulum stress, as well as improved learning and memory deficits in APP/PS1 transgenic mouse model of Alzheimer's disease (AD). However, whether tan IIA can ameliorate tau pathology and the underlying mechanism in APP/PS1 mice remains unclear. In the current study, tan IIA (15 mg/kg and 30 mg/kg) or saline was intraperitoneally administered to the 5-month-old APP/PS1 mice once daily for 4 weeks. The open-field test, novel object recognition test, Y-maze test, and Morris water maze test were performed to assess the cognitive function. Nissl staining, immunohistochemistry, TUNEL, and western blotting were conducted to explore tau hyperphosphorylation, neuronal injury, and phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt)/glycogen synthase kinase-3ß (GSK-3ß) signaling pathway. The activity of GSK-3ß, acetylcholinesterase (AChE), choline acetyltransferase (ChAT), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px), and the level of malondialdehyde (MDA) were measured using commercial kits. Our results revealed that tan IIA treatment significantly ameliorated behavioral deficits and improved spatial learning and memory ability of APP/PS1 mice. Additionally, tan IIA markedly attenuated tau hyperphosphorylation and prevented neuronal loss and apoptosis in the parietal cortex and hippocampus. Simultaneously, tan IIA reversed cholinergic dysfunction and reduced oxidative stress. Furthermore, tan IIA activated the PI3K/Akt signaling pathway and suppressed GSK-3ß. Taken together, the above findings suggested that tan IIA improves cognitive decline and tau pathology may through modulation of PI3K/Akt/GSK-3ß signaling pathway.


Assuntos
Abietanos/farmacologia , Precursor de Proteína beta-Amiloide/metabolismo , Transtornos da Memória , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Salvia miltiorrhiza , Doença de Alzheimer/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Glicogênio Sintase Quinase 3 beta/metabolismo , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/metabolismo , Camundongos , Camundongos Transgênicos , Nootrópicos/farmacologia , Transdução de Sinais/efeitos dos fármacos
5.
Mol Immunol ; 142: 22-36, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34959070

RESUMO

Activation of glial cells and neuroinflammation play an important role in the onset and development of Alzheimer's disease (AD). Triggering receptor expressed on myeloid cells 2 (TREM2) is a microglia-specific receptor in the brain that is involved in regulating neuroinflammation. However, the precise effects of TREM2 on neuroinflammatory responses and its underlying molecular mechanisms in AD have not been studied in detail. Here, we employed a lentiviral-mediated strategy to downregulation of TREM2 expression on microglia in the brain of APPswe/PS1dE9 (APP/PS1) transgenic mice and BV2 cells. Our results showed that downregulation of TREM2 significantly aggravated AD-related neuropathology including Aß accumulation, peri-plaque microgliosis and astrocytosis, as well as neuronal and synapse-associated proteins loss, which was accompanied by a decline in cognitive ability. The further mechanistic study revealed that downregulation of TREM2 expression initiated neuroinflammatory responses through toll-like receptor 4 (TLR4)-mediated mitogen-activated protein kinase (MAPK) signaling pathway and subsequent stimulating the production of pro-inflammatory cytokines in vivo and in vitro. Moreover, blockade of p38, JNK, and ERK1/2 inhibited the release of tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and interleukin-6 (IL-6) induced by Aß1-42 in TREM2-knocked down BV2 cells. Taken together, these findings indicated that TREM2 might be a potential therapeutic target for AD and other neuroinflammation-related diseases.


Assuntos
Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Glicoproteínas de Membrana/biossíntese , Doenças Neuroinflamatórias/patologia , Receptores Imunológicos/biossíntese , Receptor 4 Toll-Like/metabolismo , Doença de Alzheimer/genética , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Linhagem Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/genética , Feminino , Gliose/patologia , Sistema de Sinalização das MAP Quinases/genética , Masculino , Aprendizagem em Labirinto/fisiologia , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neuroglia/citologia , Neuroglia/patologia , Doenças Neuroinflamatórias/genética , Doenças Neuroinflamatórias/imunologia , Placa Amiloide/patologia
6.
Cell Prolif ; 54(8): e13086, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34170048

RESUMO

It is indispensable for cells to adapt and respond to environmental stresses, in order for organisms to survive. Stress granules (SGs) are condensed membrane-less organelles dynamically formed in the cytoplasm of eukaryotes cells to cope with diverse intracellular or extracellular stress factors, with features of liquid-liquid phase separation. They are composed of multiple constituents, including translationally stalled mRNAs, translation initiation factors, RNA-binding proteins and also non-RNA-binding proteins. SG formation is triggered by stress stimuli, viral infection and signal transduction, while aberrant assembly of SGs may contribute to tissue degenerative diseases. Recently, a growing body of evidence has emerged on SG response mechanisms for cells facing high temperatures, oxidative stress and osmotic stress. In this review, we aim to summarize factors affecting SGs assembly, present the impact of SGs on germ cell development and other biological processes. We particularly emphasize the significance of recently reported RNA modifications in SG stress responses. In parallel, we also review all current perspectives on the roles of SGs in male germ cells, with a particular focus on the dynamics of SG assembly.


Assuntos
Grânulos Citoplasmáticos/metabolismo , Estresse Fisiológico , Apoptose , Fatores de Iniciação em Eucariotos/metabolismo , Células Germinativas/citologia , Células Germinativas/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
7.
Exp Ther Med ; 22(1): 774, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34055073

RESUMO

B7 homolog 6 (B7-H6) was recently discovered to act as a co-stimulatory molecule. In particular, the expression of B7-H6 has been found to play an important biological role in several types of tumors. The aim of the present study was to determine the role of B7-H6 in cervical cancer. Immunohistochemistry was used to analyze the expression levels of B7-H6 in cervical precancerous and cancerous tissues. Furthermore, the expression of B7-H6 was knocked down in HeLa cells using short hairpin RNA and the effects of B7-H6 on HeLa cell proliferation, migration and invasion were determined using Cell Counting Kit-8, colony formation, wound healing and Transwell invasion assays, respectively. In addition, flow cytometry was used to analyze the levels of cell apoptosis and the cell cycle distribution. The results of the immunohistochemical staining revealed that the expression levels of B7-H6 were upregulated in cervical lesions. Furthermore, the expression levels of B7-H6 were positively associated with the clinical stage of the cervical lesions. B7-H6 knockdown suppressed the invasive, migratory and proliferative abilities of HeLa cells, and promoted G1 cell cycle arrest and apoptosis. In conclusion, the findings of the present study suggested that B7-H6 may serve as a novel oncogene and may hold promise as a potential therapeutic target for cervical cancer.

8.
Arch Gynecol Obstet ; 303(1): 207-215, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32929617

RESUMO

PURPOSE: To characterize the role of two long non-coding RNAs (lncRNAs), LINC01133 and LINC01243, in endometrial carcinoma (EC) pathogenesis. LINC01133 is an lncRNA that has been implicated in many cancers, and LINC01243 is a newly identified lncRNA identified from the NCBI GEO database. METHODS: We studied the effect of LINC01133 and LINC01243 on EC malignancy using siRNA knockdown and real-time quantitative polymerase chain reaction (RT-qPCR), flow cytometry, Annexin V-FITC/propidium iodide double staining, Transwell, and scratch invasion assays in two EC cell lines (Ishikawa and HEC-1-A cells). RESULTS: We first confirmed the partial knockdown of both LINC01133 and LINC01243 expression in Ishikawa and HEC-1-A cells using RT-qPCR. Following confirmation of lncRNA knockdown, we assessed the effect of knockdown on EC malignancy. We observed reduced EC cell proliferation using the CCK-8 assay, as well as cell cycle arrest and increased apoptosis in both EC cell lines. Furthermore, Transwell and scratch invasion assays revealed decreased migration and invasion of the two EC cell lines, respectively. CONCLUSION: We demonstrated that LINC01133 and LINC01243 expression are associated with EC development and progression. Our findings suggest a potential role for these lncRNAs as novel EC biomarkers.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , RNA Longo não Codificante/genética , Apoptose , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Progressão da Doença , Neoplasias do Endométrio/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , RNA Interferente Pequeno , Reação em Cadeia da Polimerase em Tempo Real
9.
J Neuroinflammation ; 17(1): 302, 2020 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-33054814

RESUMO

BACKGROUND: Glial activation and neuroinflammation play a crucial role in the pathogenesis and development of Alzheimer's disease (AD). The receptor for advanced glycation end products (RAGE)-mediated signaling pathway is related to amyloid beta (Aß)-induced neuroinflammation. This study aimed to investigate the neuroprotective effects of tanshinone IIA (tan IIA), a natural product isolated from traditional Chinese herbal Salvia miltiorrhiza Bunge, against Aß-induced neuroinflammation, cognitive impairment, and neurotoxicity as well as the underlying mechanisms in vivo and in vitro. METHODS: Open-field test, Y-maze test, and Morris water maze test were conducted to assess the cognitive function in APP/PS1 mice. Immunohistochemistry, immunofluorescence, thioflavin S (Th-S) staining, enzyme-linked immunosorbent assay (ELISA), real-time quantitative reverse-transcription polymerase chain reaction (qRT-PCR), and western blotting were performed to explore Aß deposition, synaptic and neuronal loss, microglial and astrocytic activation, RAGE-dependent signaling, and the production of pro-inflammatory cytokines in APP/PS1 mice and cultured BV2 and U87 cells. RESULTS: Tan IIA treatment prevented spatial learning and memory deficits in APP/PS1 mice. Additionally, tan IIA attenuated Aß accumulation, synapse-associated proteins (Syn and PSD-95) and neuronal loss, as well as peri-plaque microgliosis and astrocytosis in the cortex and hippocampus of APP/PS1 mice. Furthermore, tan IIA significantly suppressed RAGE/nuclear factor-κB (NF-κB) signaling pathway and the production of pro-inflammatory cytokines (TNF-α, IL-6, and IL-1ß) in APP/PS1 mice and cultured BV2 and U87 cells. CONCLUSIONS: Taken together, the present results indicated that tan IIA improves cognitive decline and neuroinflammation partly via inhibiting RAGE/NF-κB signaling pathway in vivo and in vitro. Thus, tan IIA might be a promising therapeutic drug for halting and preventing AD progression.


Assuntos
Abietanos/farmacologia , Anti-Inflamatórios não Esteroides/farmacologia , Mediadores da Inflamação/antagonistas & inibidores , NF-kappa B/antagonistas & inibidores , Fármacos Neuroprotetores/farmacologia , Receptor para Produtos Finais de Glicação Avançada/antagonistas & inibidores , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Linhagem Celular Tumoral , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Transgênicos , NF-kappa B/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
10.
Brain Res Bull ; 164: 136-145, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32860868

RESUMO

Tanshinone IIA (tan IIA), a key component of Salvia miltiorrhiza Bunge (Danshen), has been proven to play a significant role in suppressing inflammation. However, the molecular mechanisms underlying the anti-inflammatory properties of tan IIA against lipopolysaccharide (LPS)-induced neuroinflammation and neurotoxicity in human U87 astrocytoma cells have not been well justified. Therefore, in this study, U87 cells were pretreated with tan IIA (1, 5 and 10 µM) for 30 min, followed by stimulation with LPS for 24 h. Immunofluorescence, reverse transcription-polymerase chain reaction (RT-PCR), enzyme-linked immunosorbent assay (ELISA), and western blotting were performed to investigate the effects of tan IIA on neuroinflammatory responses. The findings demonstrated that tan IIA prevented LPS-induced cell viability decrease, inhibited U87 cells activation, and suppressed the expression of glial fibrillary acidic protein (GFAP). Furthermore, tan IIA significantly reduced the mRNA expression of interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) in LPS-stimulated U87 cells. Meanwhile, the increased protein levels of IL-1ß, TNF-α, and IL-6 in cell culture supernatants were also markedly inhibited by tan IIA. Moreover, tan IIA significantly alleviated the phosphorylation of IκBα, nuclear factor-kappa B (NF-κB), p38, and JNK induced by LPS. Additionally, tan IIA suppressed the upstream signaling adaptor molecules toll-like receptor 4 (TLR4), myeloid differentiation primary response protein 88 (MyD88), and tumor necrosis factor receptor-associated factor 6 (TRAF6). Blockade of NF-κB, p38, and JNK obviously attenuated IL-1ß, TNF-α, and IL-6 in U87 cells. In conclusion, the present results suggested that tan IIA can attenuate LPS-induced neurotoxicity and neuroinflammation partly by inhibiting TLR4/NF-κB/MAPKs signaling pathways in U87 cells.


Assuntos
Abietanos/farmacologia , Astrócitos/efeitos dos fármacos , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , NF-kappa B/metabolismo , Astrócitos/metabolismo , Linhagem Celular Tumoral , Humanos , Interleucina-6/metabolismo , Fosforilação/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
11.
Exp Cell Res ; 394(2): 112166, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32645395

RESUMO

Neuroinflammation is an important pathological feature and an early event in the pathogenesis of Alzheimer's disease (AD), which is characterized by activation of microglia and astrocytes. Low-density lipoprotein receptor-related protein 1 (LRP1) is an endocytic receptor that is abundantly expressed in neurons, microglia, and astrocytes, and plays a critical role in AD pathogenesis. There is increasing evidence to show that LRP1 regulates inflammatory responses by modulating the release of pro-inflammatory cytokines and phagocytosis. However, the effects of LRP1 on ß-amyloid protein (Aß)-induced microglial and astrocytic neuroinflammatory responses and its underlying mechanisms have not been studied in detail. In the present study, knockdown of LRP1 significantly enhanced Aß1-42-stimulated neuroinflammation by increasing the production of pro-inflammatory cytokines in both BV2 microglial cells and mouse primary astrocytes. Furthermore, it is revealed that LRP1 knockdown further led to the activation of nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinases (MAPKs) signaling pathways. The phosphorylation of IκBα, p38, and JNK was significantly up-regulated in LRP1 knockdown BV2 microglial cells and primary astrocytes. Meanwhile, LRP1 knockdown increased expression of the NF-κB p65 subunit in the nucleus while decreased its expression in the cytoplasm. Besides, the upstream signaling adaptor molecules such as toll-like receptor 4 (TLR4), myeloid differentiation primary response protein 88 (MyD88), and tumor necrosis factor receptor-associated factor 6 (TRAF6) were also further increased. Moreover, blockade of NF-κB, p38, and JNK inhibited the production of tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and interleukin-6 (IL-6) induced by the knockdown of LRP1. Taken together, these findings indicated that LRP1 as an effective therapeutic target against AD and other neuroinflammation related diseases.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Astrócitos/metabolismo , Inflamação/patologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Sistema de Sinalização das MAP Quinases , Microglia/metabolismo , NF-kappa B/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Linhagem Celular , Células Cultivadas , Citocinas/biossíntese , Citocinas/metabolismo , Técnicas de Silenciamento de Genes , Mediadores da Inflamação/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Microglia/efeitos dos fármacos , Microglia/patologia , Inibidores de Proteínas Quinases/farmacologia
12.
World Neurosurg ; 133: e31-e61, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31415895

RESUMO

BACKGROUND: The association between matrix metalloproteinase 9 (MMP-9) gene -1562C/T (rs3918242) polymorphism and the susceptibility of ischemic stroke (IS) has been investigated. However, results were ambiguous and inconsistent. Therefore, we performed this study to better assess the potential relationship between rs3918242 polymorphism and susceptibility risk of IS. METHODS: We included case-control studies concerning the relationship between the rs3918242 polymorphism and IS, and odds ratios with corresponding 95% confidence intervals were used to describe the associations. Furthermore, meta-regression analyses, heterogeneity, cumulative analyses, sensitivity analyses, and publication bias were examined. RESULTS: A total of 19 studies were included for analysis. Significant associations with the risk of IS were detected for the rs3918242 polymorphism in overall population, Asians, and whites. When available data were stratified by gender, we found a significant correlation with the risk of IS in both males and females. Further subgroup analysis by the subtypes of IS showed that the rs3918242 polymorphism was significantly correlated with the risk of patients with large artery atherosclerosis. When stratified by age, we found that the rs3918242 polymorphism was significantly correlated with the risk of IS in patients both aged ≥65 years and >65 years. Both the diabetes and the nondiabetes subgroups reached significant results, and in an analysis stratified by smoking status, an increased risk of IS was associated with smoking. CONCLUSIONS: The rs3918242 polymorphism may be a susceptible predictor of susceptibility of IS. Further large-scale studies are needed to verify the results of our findings.


Assuntos
Isquemia Encefálica/genética , Metaloproteinase 9 da Matriz/genética , Polimorfismo de Nucleotídeo Único , Adulto , Distribuição por Idade , Idoso , Aterosclerose/epidemiologia , Isquemia Encefálica/enzimologia , Isquemia Encefálica/epidemiologia , Estudos de Casos e Controles , Fumar Cigarros/epidemiologia , Comorbidade , Intervalos de Confiança , Diabetes Mellitus/epidemiologia , Etnicidade , Feminino , Predisposição Genética para Doença , Genótipo , Humanos , Embolia Intracraniana/epidemiologia , Masculino , Pessoa de Meia-Idade , Razão de Chances , Distribuição por Sexo
13.
J Cell Physiol ; 234(9): 14991-14998, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30779127

RESUMO

Targeting protein for Xenopus kinesin-like protein 2 (TPX2) is a microtubule-associated protein that plays a pivotal part in the formation of spindles. There is accumulating evidence that the expression of TPX2 is upregulated in many kinds of human cancers and that this protein is involved in the occurrence and progression of tumors. The purpose of this meta-analysis was to investigate the relationship between the overexpression of TPX2 and poor prognosis in cancer patients. A total of 18 eligible studies encompassing 3115 patients were included by searching relevant databases. Hazard ratios (HRs) or odds ratios (ORs) with 95% confidence intervals (CIs) were pooled under random-/fixed-effect models. After calculation, the results showed that patients with increased TPX2 expression had a significantly shorter overall survival (HR = 2.21; 95% CI: 1.70-2.86), and disease-free survival (HR = 2.10; 95% CI: 1.67-2.64). In addition, it was found that increased TPX2 expression was significantly associated with TNM stage (OR = 2.17; 95% CI:1.42-3.32), lymph node metastasis (OR = 2.98; 95% CI: 2.28-3.89), distant metastasis (OR = 2.25; 95% CI:1.03-4.92), and vascular invasion (OR = 2.22; 95% CI:1.26-3.91). Nevertheless, there was no significant correlation between increased expression of TPX2 and either gender, tumor differentiation, or tumor size. Thus, we can come to the conclusion that the overexpression of TPX2 is related to poor clinical outcomes and can be used as a biomarker for the prognosis of patients with cancer.

14.
Int J Biochem Cell Biol ; 107: 82-91, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30578955

RESUMO

Endoplasmic reticulum (ER) stress caused by ß-amyloid protein (Aß) may play an important role in the pathogenesis of Alzheimer disease (AD). Our previous data have indicated that tanshinone IIA (tan IIA) protected primary neurons from Aß induced neurotoxicity. To further explore the neuroprotection of tan IIA, here we study the effects of tan IIA on the ER stress response in oligomeric Aß1-42 (oAß1-42)-induced SH-SY5Y cell injury. Our data showed that tan IIA pretreatment could increase cell viability and inhibit apoptosis caused by oAß1-42. Furthermore, tan IIA markedly suppressed ER dilation and prevented oAß1-42-induced abnormal expression of glucose regulated protein 78 (GRP78), initiation factor 2α (eIF2α), activating transcription factor 6 (ATF6), as well as inhibited the activation of C/EBP homologous protein (CHOP) and c-Jun N-terminal kinase (JNK) pathways. Moreover, tan IIA ameliorated oAß1-42-induced Bcl-2/Bax ratio reduction, prevented cytochrome c translocation into cytosol from mitochondria, reduced oAß1-42-induced cleavage of caspase-9 and caspase-3, suppressed caspase-3/7 activity, and increased mitochondrial membrane potential (MMP) and ATP content. Meanwhile, oAß1-42-induced cell apoptosis and activation of ER stress can also be attenuated by the inhibitor of ER stress 4-phenylbutyric acid (4-PBA). Taken together, these data indicated that tan IIA protects SH-SY5Y cells against oAß1-42-induced apoptosis through attenuating ER stress, modulating CHOP and JNK pathways, decreasing the expression of cytochrome c, cleaved caspase-9 and cleaved caspase-3, as well as increasing the ratio of Bcl-2/Bax, MMP and ATP content. Our results strongly suggested that tan IIA may be effective in treating AD associated with ER stress.


Assuntos
Abietanos/farmacologia , Peptídeos beta-Amiloides/toxicidade , Apoptose/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , Linhagem Celular Tumoral , Citoproteção/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Resposta a Proteínas não Dobradas/efeitos dos fármacos
15.
Onco Targets Ther ; 11: 743-749, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29445289

RESUMO

OBJECTIVE: Nemo-like kinase (NLK) has been reported to play different roles in tumors. However, the role of NLK in cervical squamous cell carcinoma (CSCC) remains unknown. In this study, we explored the clinical significance including survival of NLK protein expression in CSCCs. PATIENTS AND METHODS: Immunohistochemical method was performed using tissues from 130 patients with CSCC. The associations between NLK expression and the clinicopathological factors and prognosis of CSCCs were evaluated. Statistical analyses were performed using the chi-square test, the multivariate Cox proportional hazard model, and the Kaplan-Meier method. RESULTS: Immunohistochemical staining analysis showed that NLK was localized predominately in the nucleus of the tumor cells, and increased NLK expression was detected in 71 (54.6%) of 130 patients. NLK overexpression significantly correlated with higher histological grade (P=0.001), vascular/lymphatic invasion (P=0.010), lymph node metastasis (P=0.012), and recurrence (P=0.022). Patients with elevated NLK expression had poorer overall survival (OS) and disease-free survival (DFS) (P=0.006 and P=0.004, respectively) compared with patients with decreased NLK expression. Multivariate Cox analysis demonstrated that NLK overexpression was an independent factor for OS and DFS (P=0.034 and P=0.025, respectively). CONCLUSION: NLK may be a valuable biomarker for predicting the prognosis of CSCC patients and may serve as a potential target for cancer therapy.

16.
Int J Biochem Cell Biol ; 76: 75-86, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27163530

RESUMO

Alzheimer's disease (AD) is one of the most common neurodegenerative diseases. Recently, increasing evidence suggests that intracellular ß-amyloid protein (Aß) alone plays a pivotal role in the progression of AD. Therefore, understanding the signaling pathway and proteins that control Aß internalization may provide new insight for regulating Aß levels. In the present study, the regulation of Aß internalization by p38 mitogen-activated protein kinases (MAPK) through low-density lipoprotein receptor-related protein 1 (LRP1) was analyzed in vivo. The data derived from this investigation revealed that Aß1-42 were internalized by neurons and astrocytes in mouse brain, and were largely deposited in mitochondria and lysosomes, with some also being found in the endoplasmic reticulum. Aß1-42-LRP1 complex was formed during Aß1-42 internalization, and the p38 MAPK signaling pathway was activated by Aß1-42 via LRP1. Aß1-42 and LRP1 were co- localized in the cells of parietal cortex and hippocampus. Furthermore, the level of LRP1-mRNA and LRP1 protein involved in Aß1-42 internalization in mouse brain. The results of this investigation demonstrated that Aß1-42 induced an LRP1-dependent pathway that related to the activation of p38 MAPK resulting in internalization of Aß1-42. These results provide evidence supporting a key role for the p38 MAPK signaling pathway which is involved in the regulation of Aß1-42 internalization in the parietal cortex and hippocampus of mouse through LRP1 in vivo.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Astrócitos/metabolismo , Hipocampo/metabolismo , Sistema de Sinalização das MAP Quinases , Neurônios/metabolismo , Lobo Parietal/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptores de LDL/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Feminino , Hipocampo/patologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Camundongos , Lobo Parietal/patologia , Fragmentos de Peptídeos/genética , RNA Mensageiro/metabolismo , Receptores de LDL/genética , Proteínas Supressoras de Tumor/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética
17.
Int J Biochem Cell Biol ; 64: 252-64, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25936756

RESUMO

Mounting evidence suggests that the pathological hallmarks of Alzheimer's disease (AD) are caused by the intraneuronal accumulation of beta-amyloid protein (Aß). Reuptake of extracellular Aß is believed to contribute significantly to the intraneuronal Aß pool in the early stages of AD. Published reports have claimed that the low-density lipoprotein receptor-related protein 1 (LRP1) mediates Aß1-42 uptake and lysosomal trafficking in GT1-7 neuronal cells and mouse embryonic fibroblast non-neuronal cells. However, there is no direct evidence supporting the role of LRP1 in Aß internalization in primary neurons. Our recent study indicated that p38 MAPK and ERK1/2 signaling pathways are involved in regulating α7 nicotinic acetylcholine receptor (α7nAChR)-mediated Aß1-42 uptake in SH-SY5Y cells. This study was designed to explore the regulation of MAPK signaling pathways on LRP1-mediated Aß internalization in neurons. We found that extracellular Aß1-42 oligomers could be internalized into endosomes/lysosomes and mitochondria in cortical neurons. Aß1-42 and LRP1 were also found co-localized in neurons during Aß1-42 internalization, and they could form Aß1-42-LRP1 complex. Knockdown of LRP1 expression significantly decreased neuronal Aß1-42 internalization. Finally, we identified that p38 MAPK and ERK1/2 signaling pathways regulated the internalization of Aß1-42 via LRP1. Therefore, these results demonstrated that LRP1, p38 MAPK and ERK1/2 mediated the internalization of Aß1-42 in neurons and provided evidence that blockade of LRP1 or inhibitions of MAPK signaling pathways might be a potential approach to lowering brain Aß levels and served a potential therapeutic target for AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Sistema de Sinalização das MAP Quinases , Fragmentos de Peptídeos/metabolismo , Receptores de LDL/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Linhagem Celular Tumoral , Células Cultivadas , Córtex Cerebral/citologia , Endossomos/metabolismo , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Lisossomos/metabolismo , Camundongos , Mitocôndrias/metabolismo , Neurônios , Cultura Primária de Células , Transporte Proteico
18.
Brain Res Bull ; 109: 46-53, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25290208

RESUMO

Perindopril, an angiotensin converting enzyme inhibitor, has been reported to improve learning and memory in a mouse or rat model of Alzheimer's disease (AD) induced by injection of beta-amyloid protein. However, the exact mechanism of perindopril on the cognitive deficits is not fully understood. Our previous data have indicated that perindopril improves learning and memory in a mouse model of AD induced by D-galactose (D-gal) and aluminum trichloride (AlCl3) via inhibition of acetylcholinesterase activity and oxidative stress. Whether perindopril also inhibit apoptosis to prevent cognitive decline remains unknown in mice. Therefore, the present study explored the protective effects of perindopril in the hippocampus of mice further. Perindopril (0.5 mg/kg/day) was administered intragastrically for 60 days after the mice were given a D-gal (150 mg/kg/day) and AlCl3 (10 mg/kg/day) intraperitoneally for 90 days. Then the expression of Bcl-2, Bax, Fas, FasL, caspase-3, caspase-8 and caspase-9 were analyzed by RT-PCR and western blotting in the hippocampus. Perindopril significantly decreased caspase-3 and caspase-9 activities, and elevated Bcl-2/Bax ratio in the hippocampus. However, the expression of Fas, FasL and caspase-8 did not change in the hippocampus whether treatment with d-gal and AlCl3 or perindopril. Taken together, the above findings indicated that perindopril inhibited apoptosis in the hippocampus may be another mechanism by which perindopril improves learning and memory functions in d-gal and AlCl3 treated mice.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Apoptose/efeitos dos fármacos , Hipocampo/patologia , Mitocôndrias/efeitos dos fármacos , Perindopril/farmacologia , Cloreto de Alumínio , Compostos de Alumínio/toxicidade , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Animais , Caspase 8/metabolismo , Cloretos/toxicidade , Modelos Animais de Doenças , Interações Medicamentosas , Proteína Ligante Fas/genética , Proteína Ligante Fas/metabolismo , Proteína de Domínio de Morte Associada a Fas/genética , Proteína de Domínio de Morte Associada a Fas/metabolismo , Galactose/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/ultraestrutura , Masculino , Camundongos , Mitocôndrias/metabolismo , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/etiologia , Perindopril/uso terapêutico , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
19.
Neurol Res ; 36(7): 651-8, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24620968

RESUMO

OBJECTIVES: Valsartan has been reported to reduce brain beta-amyloid protein levels and improve spatial learning in the Tg2576 transgenic mouse model of Alzheimer's disease (AD). However, the exact mechanism of neuroprotective effects of valsartan has not been properly studied especially in cholinergic function and oxidative damage, the essential factors that undergo impairment in AD. Therefore, the present study examined the effects of valsartan on memory impairment, cholinergic dysfunction, and oxidative stress in aluminum trichloride (AlCl3) and d-galactose (d-gal)-induced experimental sporadic dementia of Alzheimer's type. METHODS: Valsartan was administered intragastrically (i.g.) (20 mg/kg/day) for 60 days after mice were given AlCl3 (10 mg/kg/day) and d-gal (150 mg/kg/day) intraperitoneally (i.p.) once daily for 90 days. Then, memory function was evaluated by Morris water maze test. Acetylcholinesterase (AChE), superoxide dismutases (SOD) and glutathione peroxidase (GSH-Px) activities and malondialdehyde (MDA) level in cortex and hippocampus were also assessed with biochemical technique. RESULTS: Chronic administration of valsartan not only improved learning and memory but also restored the elevation of AChE activity induced by AlCl3 and d-gal in cortex and hippocampus. In addition, valsartan significantly restored SOD and GSH-Px activities and reduced MDA level in cortex and hippocampus indicating attenuation of oxidative stress. DISCUSSION: Our results indicate that valsartan prevents AlCl3- and d-gal-induced cognitive decline partly to restore cholinergic function and attenuate oxidative damage. These findings further support the potential of valsartan to be used in AD treatment.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Transtornos Cognitivos/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Transtornos da Memória/tratamento farmacológico , Nootrópicos/farmacologia , Tetrazóis/farmacologia , Valina/análogos & derivados , Acetilcolinesterase/metabolismo , Cloreto de Alumínio , Compostos de Alumínio , Animais , Córtex Cerebral/fisiopatologia , Cloretos , Transtornos Cognitivos/fisiopatologia , Demência , Modelos Animais de Doenças , Galactose , Glutationa Peroxidase/metabolismo , Hipocampo/fisiopatologia , Malondialdeído/metabolismo , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/fisiopatologia , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Distribuição Aleatória , Superóxido Dismutase/metabolismo , Valina/farmacologia , Valsartana
20.
Pharmacol Biochem Behav ; 114-115: 31-6, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24201055

RESUMO

Preclinical and clinical studies indicate involvement of renin angiotensin system (RAS) in memory functions. However, exact role of RAS in cognition is still ambiguous. The present study investigated the effects of perindopril on dementia of Alzheimer's type induced by d-galactose (d-gal) and aluminum trichloride (AlCl3). Perindopril, an angiotensin converting enzyme inhibitor, was administered intragastrically (0.5mg/kg/day) for 60days after mice were given d-gal (150mg/kg/day) and AlCl3 (10mg/kg/day) intraperitoneally for 90days. Then, memory function was evaluated by Morris water maze test. The biochemical studies were conducted in cerebral cortex and hippocampus of mouse brain after the behavioral studies. d-Gal and AlCl3 caused significant memory impairment along with significant elevation of acetylcholinesterase (AChE) activity in cerebral cortex and hippocampus. Further, a significant reduction of superoxide dismutases (SOD) and glutathione peroxidase (GSH-Px) activities, and elevation of malondialdehyde (MDA) level in cerebral cortex and hippocampus were observed. Perindopril not only improved cognitive impairment but also restored the elevation of AChE activity induced by d-gal and AlCl3. In addition, perindopril significantly increased SOD and GSH-Px activities, reduced MDA level in cerebral cortex and hippocampus. Taken together, the above findings indicate that perindopril improves learning and memorizing probably by restoring cholinergic function and attenuating oxidative damage.


Assuntos
Compostos de Alumínio/administração & dosagem , Cloretos/administração & dosagem , Inibidores da Colinesterase/farmacologia , Transtornos Cognitivos/tratamento farmacológico , Galactose/administração & dosagem , Estresse Oxidativo/efeitos dos fármacos , Perindopril/uso terapêutico , Acetilcolinesterase/metabolismo , Cloreto de Alumínio , Compostos de Alumínio/farmacologia , Animais , Cloretos/farmacologia , Transtornos Cognitivos/induzido quimicamente , Galactose/farmacologia , Glutationa Peroxidase/metabolismo , Masculino , Malondialdeído/metabolismo , Camundongos , Superóxido Dismutase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA