Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38978503

RESUMEN

Lung adenocarcinoma (LUAD) remains a predominant cause of cancer-related mortality globally, underscoring the urgency for targeted therapeutic strategies. The specific role and impact of the SEC61 translocon gamma subunit (SEC61G) in LUAD progression and metastasis remain largely unexplored. In this study, we use a multifaceted approach, combining bioinformatics analysis with experimental validation, to elucidate the pivotal role of SEC61G and its associated molecular mechanisms in LUAD. Our integrated analyses reveal a significant positive correlation between SEC61G expression and the glycolytic activity of LUAD, as evidenced by increased fluorodeoxyglucose (FDG) uptake on positron emission tomography (PET)/CT scans. Further investigations show the potential influence of SEC61G on metabolic reprogramming, which contributes to the immunosuppressive tumor microenvironment (TME). Remarkably, we identify a negative association between SEC61G expression levels and the infiltration of critical immune cell populations within the TME, along with correlations with immune checkpoint gene expression and tumor heterogeneity scores in LUAD. Functional studies demonstrate that SEC61G knockdown markedly inhibits the migration of A549 and H2030 LUAD cells. This inhibitory effect is accompanied by a significant downregulation of key regulators of tumor progression, including hypoxia-inducible factor-1 alpha (HIF-1α), lactate dehydrogenase A, and genes involved in the epithelial-mesenchymal transition pathway. In conclusion, our comprehensive analyses position SEC61G as a potential prognostic biomarker intricately linked to glycolytic metabolism, the EMT pathway, and the establishment of an immune-suppressive phenotype in LUAD. These findings underscore the potential of SEC61G as a therapeutic target and predictive marker for immunotherapeutic responses in LUAD patients.

2.
Mol Cell Biochem ; 478(7): 1415-1425, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36348200

RESUMEN

Cognitive dysfunction has been regarded as a complication of diabetes. Melatonin (MLT) shows a neuroprotective effect on various neurological diseases. However, its protective effect on cortical neurons in high glucose environment has not been reported. Our present study aims to observe the protective effect of melatonin on rat cortical neurons and its relationship with autophagy in high glucose environment. The rat primary cortical neurons injury model was induced by high glucose. The CCK-8, flow cytometry, Western blot and immunofluorescence methods were used to examine the cell viability, apoptosis rate and proteins expression. Our results showed that there were no differences in cell viability, apoptosis rate, and protein expression among the control, MLT and mannitol group. The cell viability of the glucose group was significantly lower than that of the control group, and the apoptosis rate of the glucose group was significantly higher than that of the control group. Compared with the glucose group, the glucose + melatonin group showed a significant increase in cell viability and a notable decrease in apoptosis rate. Melatonin concentration of 0.1-1 mmol/L can significantly alleviate the injury of cortical neurons caused by high glucose. Compared with the control group, the glucose group showed a significant reduction of B-cell lymphoma 2 (Bcl-2) protein expression, while remarkable elevations of Bcl2-associated X protein (Bax), cleaved Caspase-3, coiled-coil, myosin-like Bcl2-interacting protein (Beclin-1) and microtubule-associated protein 1 light chain-3B type II (LC3B-II) levels. The neurons pre-administered with melatonin obtained significantly reversed these changes induced by high glucose. The phosphorylation levels of protein kinase B (Akt), mechanistic target of rapamycin kinase (mTOR) and Unc-51 like autophagy activating kinase 1(ULK1) were decreased in the glucose group compared with the control group, whereas significant increase were observed in the glucose + MLT group, compared with the glucose group. These data indicated that melatonin has a neuroprotective effect on cortical neurons under high glucose environment, which may work by activating Akt/mTOR/ULK1 pathway and may be deeply associated with the downregulation of autophagy.


Asunto(s)
Melatonina , Fármacos Neuroprotectores , Ratas , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Melatonina/farmacología , Melatonina/metabolismo , Fármacos Neuroprotectores/farmacología , Apoptosis , Glucosa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Autofagia , Neuronas/metabolismo
3.
Int J Mol Sci ; 24(6)2023 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-36982952

RESUMEN

Nucleolar and spindle-associated protein 1 (NUSAP1) is a potential molecular marker and intervention target for glioblastoma (GBM). In this study, we aim to investigate upstream regulatory lncRNAs and miRNAs of NUSAP1 through both experimental and bioinformatic methods. We screened upstream lncRNAs and miRNAs of NUSAP1 through multiple databases based on ceRNA theory. Then, in vitro and in vivo experiments were performed to elucidate the relevant biological significance and regulatory mechanism among them. Finally, the potential downstream mechanism was discussed. LINC01393 and miR-128-3p were screened as upstream regulatory molecules of NUSAP1 by TCGA and ENCORI databases. The negative correlations among them were confirmed in clinical specimens. Biochemical studies revealed that overexpression or knockdown of LINC01393 respectively enhanced or inhibited malignant phenotype of GBM cells. MiR-128-3p inhibitor reversed LINC01393 knockdown-mediated impacts on GBM cells. Then, dual-luciferase reporter assay and RNA immunoprecipitation assay were conducted to validate LINC01393/miR-128-3p/NUSAP1 interactions. In vivo, LINC01393-knockdown decreased tumor growth and improved mice survival, while restoration of NUSAP1 partially reversed these effects. Additionally, enrichment analysis and western blot revealed that the roles of LINC01393 and NUSAP1 in GBM progression were associated with NF-κB activation. Our findings showed that LINC01393 sponged miR-128-3p to upregulate NUSAP1, thereby promoting GBM development and progression via activating NF-κB pathway. This work deepens understanding of GBM mechanisms and provides potential novel therapeutic targets for GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , MicroARNs , ARN Largo no Codificante , Animales , Ratones , Glioblastoma/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/uso terapéutico , FN-kappa B/metabolismo , Neoplasias Encefálicas/metabolismo , MicroARNs/metabolismo , Proliferación Celular/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica
4.
Mol Med ; 28(1): 142, 2022 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-36447154

RESUMEN

BACKGROUND: Activated microglia play a key role in initiating the inflammatory cascade following ischemic stroke and exert proinflammatory or anti-inflammatory effects, depending on whether they are polarized toward the M1 or M2 phenotype. The present study investigated the regulatory effect of icaritin (ICT) on microglial polarization in rats after cerebral ischemia/reperfusion injury (CI/RI) and explored the possible anti-inflammatory mechanisms of ICT. METHODS: A rat model of transient middle cerebral artery occlusion (tMCAO) was established. Following treatment with ICT, a G protein-coupled estrogen receptor (GPER) inhibitor or an extracellular signal-regulated kinase (ERK) inhibitor, the Garcia scale and rotarod test were used to assess neurological and locomotor function. 2,3,5-Triphenyltetrazolium chloride (TTC) and Fluoro-Jade C (FJC) staining were used to evaluate the infarct volume and neuronal death. The levels of inflammatory factors in the ischemic penumbra were evaluated using enzyme-linked immunosorbent assays (ELISAs). In addition, western blotting, immunofluorescence staining and quantitative PCR (qPCR) were performed to measure the expression levels of markers of different microglial phenotypes and proteins related to the GPER-ERK-nuclear factor kappa B (NF-κB) signaling pathway. RESULTS: ICT treatment significantly decreased the cerebral infarct volume, brain water content and fluorescence intensity of FJC; improved the Garcia score; increased the latency to fall and rotation speed in the rotarod test; decreased the levels of interleukin-1 beta (IL-1ß), tumor necrosis factor-alpha (TNF-α), Iba1, CD40, CD68 and p-P65-NF-κB; and increased the levels of CD206 and p-ERK. U0126 (an inhibitor of ERK) and G15 (a selective antagonist of GPER) antagonized these effects. CONCLUSIONS: These findings indicate that ICT plays roles in inhibiting the inflammatory response and achieving neuroprotection by regulating GPER-ERK-NF-κB signaling and then inhibiting microglial activation and M1 polarization while promoting M2 polarization, which provides a new therapeutic for against cerebral ischemic stroke.


Asunto(s)
Accidente Cerebrovascular Isquémico , FN-kappa B , Animales , Ratas , Quinasas MAP Reguladas por Señal Extracelular , Microglía , Enfermedades Neuroinflamatorias , Infarto Cerebral , Estrógenos , Transducción de Señal
5.
Acta Biochim Biophys Sin (Shanghai) ; 55(3): 417-425, 2022 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-36305723

RESUMEN

Glioblastoma multiforme (GBM) is a highly vascularized malignant brain tumor. Our previous study showed that prostate-specific membrane antigen (PSMA) promotes angiogenesis of GBM. However, the specific mechanism underlying GBM-induced PSMA upregulation remains unclear. In this study, we demonstrate that the GBM-secreted cytokine phosphoprotein 1 (SPP1) can regulate the expression of PSMA in human umbilical vein endothelial cells (HUVECs). Our mechanistic study further reveals that SPP1 regulates the expression of PSMA through the transcription factor HIF1α. Moreover, SPP1 promotes HUVEC migration and tube formation. In addition, HIF1α knockdown reduces the expression of PSMA in HUVECs and blocks the ability of SPP1 to promote HUVEC migration and tube formation. We further confirm that SPP1 is abundantly expressed in GBM, is associated with poor prognosis, and has high clinical diagnostic value with considerable sensitivity and specificity. Collectively, our findings identify that the GBM-secreted cytokine SPP1 upregulates PSMA expression in endothelial cells via the transcription factor HIF1α, providing insight into the angiogenic process and promising candidates for targeted GBM therapy.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Masculino , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Glioblastoma/genética , Glioblastoma/irrigación sanguínea , Glioblastoma/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Patológica/metabolismo , Osteopontina/metabolismo , Factores de Transcripción/metabolismo
6.
J Cell Mol Med ; 25(10): 4826-4834, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33793070

RESUMEN

Cardiac conduction regulatory RNA (CCRR) is down-regulated in the pathogenesis of heart failure (HF), which accordingly suppresses cardiac conduction while promoting arrhythmogenicity. Meanwhile, CX43 was reported to play a role in the pathogenesis of metastatic breast cancer and melanoma brain colonization. In this study, we studied the role of long non-coding RNA CCRR and its interaction with CX43 in brain metastasis of breast cancer. Breast cancer patients were grouped according to the metastasis status. Real-time PCR and IHC assay were used to measure the expression of lncRNA-CCRR and CX43 in patients. Western blot was conducted to observe the effect of lncRNA-CCRR on the expression of CX43 in MDA-MB-231BR and BT-474BR cells. Compared with the non-metastasis group, the mRNA expression of tissue lncRNA-CCRR, cerebrospinal fluid (CSF) lncRNA-CCRR, tissue CX43 and tissue protein expression of CX43 were both evidently up-regulated in metastasis patients, especially in patients with brain metastasis. The expression of lncRNA-CCRR was positively correlated with the up-regulated expression of CX43. Moreover, CX43 expression was significantly lower in MDA-MB-231WT cells compared with that in MDA-MB-231BR cells. Also, the overexpression of lncRNA-CCRR evidently increased dye transfer rate from astrocytes to MDA-MB-231BR/BT-474BR cells but reduced lncRNA-CCRR expression and suppressed the transmigration of MDA-MB-231BR/BT-474BR cells in a blood-brain barrier (BBB) model. In this study, we demonstrated that the presence of lncRNA-CCRR could up-regulate the expression of CX43, which promoted gap junction formation in brain metastasis of breast cancer. Accordingly, the communication between breast cancer cells and astrocytes was also promoted.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/patología , Conexina 43/metabolismo , Regulación Neoplásica de la Expresión Génica , ARN Largo no Codificante/genética , Apoptosis , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Movimiento Celular , Proliferación Celular , Conexina 43/genética , Femenino , Humanos , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas
7.
J Cell Mol Med ; 24(22): 13010-13019, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32996285

RESUMEN

Long noncoding RNAs have key roles in glioma progression. However, the function and mechanisms of action of the long noncoding RNA, LINC00346, in glioma remain unclear. In our study, we observed that LINC00346 levels were increased in glioma tissue samples, and according to Gene Expression Profiling Interactive Analysis, its levels were related to disease-free survival and overall survival rates, suggesting that a high level of LINC00346 expression corresponds to a poor prognosis. We next confirmed the high levels of LINC00346 expression in glioma tissues and cell lines and showed that LINC00346 knockdown suppressed glioma cell proliferation, migration and invasion; promoted apoptosis; and delayed tumour growth. Moreover, the oncogenic function of LINC00346 may be explained, in part, by the down-regulation of miR-340-5p and the de-repression of ROCK1. We showed that LINC00346 may function as a competing endogenous RNA of miR-340-5p, thereby de-repressing ROCK1. This study revealed a new regulatory network in glioma and identified potential therapeutic targets for this cancer.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , ARN Largo no Codificante/genética , Quinasas Asociadas a rho/metabolismo , Animales , Apoptosis , Astrocitos/metabolismo , Carcinogénesis/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Biología Computacional , Supervivencia sin Enfermedad , Regulación hacia Abajo , Humanos , Ratones , Ratones Desnudos , MicroARNs/genética , Invasividad Neoplásica , Pronóstico , Activación Transcripcional , Regulación hacia Arriba , Quinasas Asociadas a rho/genética
8.
Mol Carcinog ; 59(4): 390-398, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32037613

RESUMEN

Brain metastasis is a leading cause of death worldwide, but the mechanism involved remains unclear. Stromal cell-derived factor-1 (SDF-1)/C-X-C motif chemokine receptor 4 (CXCR4) signaling has been reported to induce the directed metastasis of cancers, and adenosine A2A receptor activation suppresses the SDF-1/CXCR4 interaction. However, whether A2A receptor activation implicates the SDF-1/CXCR4 signaling pathway and thus modulates brain metastasis remains unclear. In this study, Western blot was performed to evaluate the protein levels. Cell invasion and migration assays were used to estimate the metastasis ability of PC-9 cells. The viability of cells was demonstrated by lactate dehydrogenase and cell proliferation assays. And the findings in vitro were further identified in nude mice. Notably, adenosine A2A receptor activation inhibited the proliferation and viability of PC-9 cells and thus suppressed the brain metastasis. A2A receptor stimulation protected the function of blood-brain barrier (BBB). The suppression of brain metastasis and the protection of BBB by A2A receptor relied on SDF-1/CXCR4 signaling, and treatment using A2A receptor agonist and CXCR4 antagonist protected the nude mice from malignancy metastasis in vivo. Adenosine A2A receptor activation suppressed the brain metastasis by implicating the SDF-1/CXCR4 axis and protecting the BBB.


Asunto(s)
Agonistas del Receptor de Adenosina A2/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Neoplasias Encefálicas/prevención & control , Quimiocina CXCL12/metabolismo , Neoplasias Pulmonares/prevención & control , Receptor de Adenosina A2A/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Bencilaminas , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/secundario , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Ciclamas , Compuestos Heterocíclicos/farmacología , Humanos , Neoplasias Pulmonares/patología , Ratones Endogámicos BALB C , Ratones Desnudos , Fenetilaminas/farmacología , Receptores CXCR4/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
9.
J Neurooncol ; 146(3): 513-521, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32020481

RESUMEN

PURPOSE: Glioma is the most common malignant primary tumor in the central nervous system (CNS). KIF3C, a motor protein of the kinesin superfamily, is highly expressed in the CNS. Although KIF3C has been identified as a potential therapeutic target in malignant cancers, the expression and function of KIF3C in glioma remains unclear. METHODS: The clinical characteristics of 53 patients with graded glioma (WHO I-IV) were analyzed in this study. The expression of KIF3C in glioma was evaluated by immunohistochemistry (IHC). Survival analysis was compared between higher and lower KIF3C expression groups. Data regarding the expression of KIF3C and survival analysis were also confirmed using the database from The Cancer Genome Atlas (TCGA). The potential mechanism of the regulation of tumor growth by KIF3C was investigated by an analysis of the public database from Oncomine. RESULTS: Expression of the KIF3C protein was higher in the low-grade glioma (LGG) group (n = 20) than that in the high-grade glioma (HGG) group (n = 33) (P < 0.05). Glioma patients with higher expression of KIF3C had longer survival time (P < 0.05). The subgroup analysis showed that higher KIF3C expression predicted longer survival time in the LGG group (P < 0.05). These clinical results were consistent with those in the TCGA database. Bioinformatics analysis showed that the KIF3C mRNA expression was upregulated significantly in response to PI3K/AKT/mTOR pathway inhibition. CONCLUSION: This study demonstrated that KIF3C might inhibit glioma growth to prolong survival time by regulating the PI3K/AKT/mTOR pathway, providing a potential therapeutic target in glioma.


Asunto(s)
Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioma/diagnóstico , Glioma/metabolismo , Cinesinas/metabolismo , Transducción de Señal , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fosfatidilinositol 3-Quinasas/metabolismo , Pronóstico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Análisis de Supervivencia , Serina-Treonina Quinasas TOR/metabolismo
10.
Environ Toxicol ; 35(6): 697-706, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31995275

RESUMEN

Here, we aim to explore whether tribbles pseudokinase 3 (TRIB3) enhances glioma cell stemness. TRIB3 was overexpressed in glioma tissues and cell-formed spheres, positively correlated with the size and grade. Additionally, TRIB3 expression displayed a negative correlation with the overall survival rate of glioma patients. Moreover, TRIB3 knockdown reduced the stemness of nonadherent spheres, evident by the decreased sphere-forming ability, stemness master expression, and ALDH1 activity, while TRIB3 overexpression enhanced the stemness of adherent cells, which was rescued by ß-catenin knockdown. Mechanistically, TRIB3 activated ß-catenin signaling via physically interacting with ß-catenin. This study suggests that the TRIB3-ß-catenin interaction is responsible for glioma cell stemness.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Glioma/patología , Células Madre Neoplásicas/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Represoras/metabolismo , beta Catenina/metabolismo , Adulto , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glioma/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Células Madre Neoplásicas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Represoras/genética , Vía de Señalización Wnt
11.
BMC Cancer ; 18(1): 1256, 2018 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-30558666

RESUMEN

OBJECTIVE: Exhibiting high consistence in sequence and structure, S100 family members are interchangeable in function and they show a wide spectrum of biological processes, including proliferation, apoptosis, migration, inflammation and differentiation and the like. While the prognostic value of each individual S100 in ovarian cancer is still elusive. In current study, we investigated the prognostic value of S100 family members in the ovarian cancer. METHODS: We used the Kaplan Meier plotter (KM plotter) database, in which updated gene expression data and survival information are from 1657 ovarian cancer patients, to assess the relevance of individual S100 family mRNA expression to overall survival in various ovarian cancer subtypes and different clinicopathological features. RESULTS: It was found that high expression of S100A2 (HR = 1.18, 95%CI: 1.04-1.34, P = 0.012), S100A7A (HR = 1.3, 95%CI: 1.04-1.63, P = 0.02),S100A10 (HR = 1.2, 95%CI: 1.05-1.38, P = 0.0087),and S100A16 (HR = 1.23, 95%CI: 1-1.51, P = 0.052) were significantly correlated with worse OS in all ovarian cancer patients, while the expression of S100A1 (HR = 0.87, 95%CI: 0.77-0.99, P = 0.039), S100A3 (HR = 0.83, 95%CI: 0.71-0.96, P = 0.0011), S100A5 (HR = 0.84, 95%CI: 0.73-0.97, P = 0.017), S100A6 (HR = 0.84, 95%CI: 0.72-0.98, P = 0.024), S100A13 (HR = 0.85, 95%CI:0.75-0.97, P = 0.014) and S100G (HR = 0.86, 95%CI: 0.74-0.99, P = 0.041) were associated with better prognosis. Furthermore, we assessed the prognostic value of S100 expression in different subtypes and the clinicopathological features, including pathological grades, clinical stages and TP53 mutation status, of ovarian cancer patients. CONCLUSION: Comprehensive understanding of the S100 family members may have guiding significance for the diagnosis and outcome of ovarian cancer patients.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Proteínas S100/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Mutación , Clasificación del Tumor , Pronóstico , Análisis de Supervivencia , Proteína p53 Supresora de Tumor/genética
12.
Future Oncol ; 13(19): 1711-1719, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28592145

RESUMEN

BACKGROUND: Cytochrome c oxidase subunit VB (COX5B), a subunit of mammalian COX, takes roles in COX assembling and functions. Online database predicts high COX5B transcription may be associated with worse disease-free survival (DFS). However, the clinical implications of COX5B in breast cancer remain unclear. METHODS: We carried out immunohistochemistry on tissue microarrays of 244 patients with invasive ductal breast carcinoma to detected COX5B expression. RESULTS: Our results suggest that COX5B protein level might be associated with tumor size. COX5B overexpression indicated a worse DFS (p < 0.05) in breast cancer. Furthermore, high COX5B expression may act as an independent factor for worse DFS in breast cancer. CONCLUSIONS: Cumulatively, our findings suggest that COX5B might serve as an important prognostic factor for breast cancer.


Asunto(s)
Biomarcadores de Tumor , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Complejo IV de Transporte de Electrones/genética , Expresión Génica , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/patología , Complejo IV de Transporte de Electrones/metabolismo , Femenino , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Clasificación del Tumor , Metástasis de la Neoplasia , Estadificación de Neoplasias , Pronóstico , Análisis de Supervivencia
13.
Tumour Biol ; 37(1): 1279-87, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26289846

RESUMEN

TIM50 is an essential component of TIM23 complex and involved in protein translocating into the inner mitochondrial membrane. Here, we found that TIM50 was increased in breast cancer cells by SILAC. However, its biological functions and molecular mechanisms in breast cancer are poorly understood. To gain insight into the functions of TIM50 in breast cancer, we constructed two stably transfected cell lines and examined TIM50 expression in tissue samples. Our data showed that TIM50 expression was increased in breast cancer. The stable suppression of TIM50 expression through lentivirus-mediated shRNA was shown to inhibit the abilities of cancer cell proliferation and induce apoptosis. What is more, depletion of TIM50 could decrease mitochondrial membrane potential, which may be associated with cell viability. Taken together, our findings reveal a new role for TIM50 in regulating cell proliferation and apoptosis through decreasing mitochondrial membrane potential in breast cancer cell and suggest that TIM50 might be a potential target for controlling breast cancer progression.


Asunto(s)
Apoptosis , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , Proteínas de Transporte de Membrana/metabolismo , Muerte Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Femenino , Citometría de Flujo , Silenciador del Gen , Humanos , Potenciales de la Membrana , Mitocondrias/patología , Membranas Mitocondriales/metabolismo , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Transporte de Proteínas , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
Heliyon ; 10(9): e30727, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38774095

RESUMEN

Background: The FXYD domain-containing ion transport regulator 5 (FXYD5) gene is a cancer promoter. However, evidence for an association between FXYD5 and various types of cancer is still lacking. Using multi-omics bioinformatics, our study aimed to reveal the expression distribution, prognostic value, immune infiltration correlation, and molecular functions of FXYD5. Methods: Using pan-cancer multi-omics data (including The Cancer Genome Atlas, PrognoScan, Gene Expression Profiling Interactive Analysis, cBioPortal, Gene Expression Omnibus, TIMER and scTIME Portal), we assessed the differences in the expression and prognostic value of FXYD5 in malignant tumors. Furthermore, at the single-cell level, we analyze the expression distribution of FXYD5 across different cell types within the tumor microenvironment, and its relationship with the immune microenvironment. Finally, focusing on ovarian cancer, we conducted preliminary validation of the above findings using cell and molecular biology techniques. Results: Our results indicated that FXYD5 was up-regulated in various tumor types and was positively associated with tumor progression. We also revealed that FXYD5 was ubiquitously expressed in microenvironmental cells at the single-cell level, and its upregulation was associated with enhanced immune infiltration, cancer-associated fibroblast infiltration, and dysfunction of tumor-infiltrating cytotoxic T lymphocyte. Additionally, its expression was positively correlated with immune checkpoint genes, DNA mismatch repair genes, MSI (microsatellite instability) and TMB (tumor mutational burden) across various cancers. Its higher expression in cytotoxic T lymphocytes attenuated its ability to predict patient survival with PD-L1 (programmed death-ligand 1) blockade therapy, and FXYD5 was found to be a potential regulator of tumor immune escape and resistance to cancer immunotherapies. Based on GSEA (gene set enrichment analysis) and experimental verification, FXYD5 activated TGF-ß/SMAD signaling and drove EMT (epithelial-mesenchymal transition) to promote ovarian cancer progression. Conclusion: In summary, our study revealed that FXYD5-TGFß axis may coregulate the interaction between tumors, CAFs (carcinoma-associated fibroblasts) and immune cells to reshape the tumor immune microenvironment and promote tumorigenesis and tumor progression. Thus, FXYD5 could be used as an immune-related biomarker for diagnosing and predicting the prognosis of multiple cancer types. Therefore, our findings suggest that targeting FXYD5 in TME (tumor microenvironment) may be a promising therapeutic strategy.

15.
Transl Oncol ; 28: 101616, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36621073

RESUMEN

Brain metastases (BMs) of lung cancer are common malignant intracranial tumours associated with severe neurological symptoms and an abysmal prognosis. Prostate-specific membrane antigen (PSMA) has been reported to express significantly in a variety of solid tumours. However, the clinical applications of 68Ga-PSMA PET/CT and the mechanism of PSMA expression in patients with BMs of lung cancer have rarely been reported. Experiments with 68Ga-PSMA PET/CT and immunohistochemical staining were conducted to evaluate the expression of PSMA from seven patients with BMs of lung cancer who accepted surgical treatment in Fudan University Shanghai Cancer Center between October 2020 and October 2021. The mechanism of PSMA expression in BMs of lung cancer was explored by using single-cell RNA sequencing. The median maximum standardized uptake value (SUVmax) in BMs was higher than that in primary lung cancer (8.6 ± 2.8 vs. 3.6 ± 1.3, P < 0.01). The mean SUVmax in BMs was 1.76-fold higher than that in the liver, which indicated the potential of PSMA radioligand therapy (PSMA-RLT) for BMs. BMs showed intense PSMA staining, while normal lung tissue had no PSMA staining and there was only faint primary lung cancer staining by immunohistochemistry (IHC). Single-cell RNA sequencing (scRNA-seq) analysis found that PSMA was mainly expressed in oligodendrocytes of BMs, whereas it was expressed at lower levels in solid cells of lung cancer. PSMA expression in oligodendrocytes might be regulated by the factors ATF3 and NR4A1, which were associated with ER stress.

16.
Eur J Pharmacol ; 941: 175492, 2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36610684

RESUMEN

N-methyl-D-aspartate (NMDA) receptors are key signaling molecules that mediate excitotoxicity during cerebral ischemia. GluN2A-containing NMDA receptors, which are mostly located in the intrasynaptic region, mediate normal physiological processes and promote neuronal survival. GluN2B-containing NMDA receptors, which are mostly located in the extrasynaptic region, mediate excitotoxicity injury and promote neuronal death during ischemia. This study investigated the ability of icaritin (ICT) to protect against cerebral ischemia‒reperfusion injury (CI/RI) by regulating GluN2B-containing NMDA receptors through extracellular signaling regulatory kinases/death associated protein kinase 1 (ERK/DAPK1) signaling. A rat CI/RI model was established by transient middle cerebral artery occlusion (tMCAO). Following treatment with ICT and the ERK-specific inhibitor U0126, cerebral infarction, neurological function, and excitotoxicity-related molecule expression were assessed 24 h after reperfusion. ICT treatment significantly decreased cerebral infarct volume, improved neurological function, and regulated NMDA receptor subtype expression and ERK/DAPK1 signaling activation. The ability of ICT to increase GluN2A and postsynaptic density protein 95 (PSD95) mRNA and protein expression, inhibit GluN2B expression, and regulate DAPK1 activation was reversed after administration of the ERK-specific inhibitor U0126. These data indicated that ICT inhibited excitotoxicity injury and exerted a protective effect against CI/RI that was likely mediated by increased ERK signaling pathway activation and regulation of extrasynaptic and intrasynaptic NMDA receptor function, providing a new therapeutic target for ischemic encephalopathy.


Asunto(s)
Isquemia Encefálica , Daño por Reperfusión , Ratas , Animales , Receptores de N-Metil-D-Aspartato/metabolismo , Neuronas , Transducción de Señal , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo
17.
Adv Sci (Weinh) ; 9(12): e2105672, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35199941

RESUMEN

Triple-negative breast cancer is the most aggressive subtype of breast cancer and the incidence of depression in breast cancer patients is high, which leading to worse survival and increased risk of recurrence. The effect of antidepressants on breast cancer patients remains contradictory, which might be due to variations in antidepression targets. Therefore, there is significant value to explore the antitumor potential of antidepressants and discover new therapeutic targets for breast patients. The authors screen antidepressant-related oncogenes or suppressors by using siRNAs. After combining functional experiments with online database analysis, 5-hydroxytryptamine receptor 1A (HTR1A is selected with antitumor potential in breast cancer cells in vivo and in vitro. RNA-seq analysis and coimmunoprecipitation assays indicate that HTR1A interacts with TRIM21 and PSMD7 to inhibit the degradation of TßRII through the ubiquitin-proteasome pathway, thereby inhibiting the transforming growth factor-ß (TGF-ß) canonical and noncanonical pathway. In addition, HTR1A is an independent predictive factor for breast cancer patients. The combined treatment of HTR1A agonists with demethylation drugs may significantly improve patient survival. It is of great significance to clarify the function and mechanism of the depression-related gene HTR1A in breast cancer, which might provide a new approach for triple-negative breast cancer patients.


Asunto(s)
Receptor de Serotonina 5-HT1A , Neoplasias de la Mama Triple Negativas , Humanos , Pronóstico , Receptor de Serotonina 5-HT1A/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
18.
Clin Transl Med ; 12(11): e1101, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36336787

RESUMEN

BACKGROUND: Brain malignancies encompass gliomas and brain metastases originating from extracranial tumours including lung cancer. Approximately 50% of patients with lung adenocarcinoma (LUAD) will eventually develop brain metastases. However, the specific characteristics of gliomas and lung-to-brain metastases (LC) are largely unknown. METHODS: We applied single-cell RNA sequencing to profile immune and nonimmune cells in 4 glioma and 10 LC samples. RESULTS: Our analysis revealed that tumour microenvironment (TME) cells are present in heterogeneous subpopulations. LC reprogramed cells into immune suppressed state, including microglia, macrophages, endothelial cells, and CD8+ T cells, with unique cell proportions and gene signatures. Particularly, we identified that a subset of macrophages was associated with poor prognosis. ROS (reactive oxygen species)-producing neutrophils was found to participant in angiogenesis. Furthermore, endothelial cells participated in active communication with fibroblasts. Metastatic epithelial cells exhibited high heterogeneity in chromosomal instability (CIN) and cell population. CONCLUSIONS: Our findings provide a comprehensive understanding of the heterogenicity of the tumor microenvironment and tumour cells and it will be crucial for successful immunotherapy development for brain metastasis of lung cancer.


Asunto(s)
Neoplasias Encefálicas , Glioma , Neoplasias Pulmonares , Humanos , Linfocitos T CD8-positivos/patología , Células Endoteliales/patología , Glioma/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Análisis de Secuencia de ARN , Microambiente Tumoral/genética
19.
Front Genet ; 13: 975381, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36105075

RESUMEN

Phosphoserine aminotransferase 1 (PSAT1) may be an oncogene that plays an important role in various cancer types. However, there are still many gaps in the expression of PSAT1 gene and its biological impact in different types of tumors. Here, we performed an integrated pan-cancer analysis to explore the potential molecular mechanisms of PSAT1 in cancers. We found that most human tumors express higher levels of PSAT1 than normal tissues, and that higher PSAT1 expression is associated with worse prognosis in Lung adenocarcinoma (LUAD), Pan-kidney cohort (KIPAN) and breast invasive carcinoma (BRCA), etc. In BRCA cases, the prognosis of patients with altered PSAT1 was worse than that of patients without alteration. In addition, PSAT1 hypermethylation is associated with T cell dysfunction and shortened survival time in BRCA. The Gene Set Enrichment Analysis (GSEA) analysis showed that PSAT1 can be enriched into the classic signaling pathways of cancer such as mTORC1 signaling, MYC targets and JAK STAT3. Further analysis demonstrated that PSAT1 was enriched in immune related signaling pathways in LUAD and BRCA. The results of immunoassay showed that PSAT1 was associated with immune cell infiltration in multiple cancer species. Furthermore, expression of PSAT1 was correlated with both tumor mutational burden (TMB) and microsatellite instability (MSI) in BRCA. Additionally, a remarkable correlation was found between PSAT1 expression and TMB in LUAD, and the expression of PSAT1 was negatively correlated with the Tumor Immune Dysfunction and Exclusion (TIDE) value, suggesting a good effect of immunotherapy. Together, these data suggest that PSAT1 expression is associated with the clinical prognosis, DNA methylation, gene mutations, and immune cell infiltration, contributing to clarify the role of PSAT1 in tumorigenesis from a variety of perspectives. What's more, PSAT1 may be a new biomarker for survival and predicting the efficacy of immunotherapy for LUAD and BRCA.

20.
Transl Oncol ; 22: 101455, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35598384

RESUMEN

OBJECTIVES: Brain metastases from lung adenocarcinoma cause significant patient mortality. This study aims to evaluate the role of preoperative Neutrophil-to-Lymphocyte ratio (preNLR) in predicting the survival and prognosis of Lung adenocarcinoma (LUAD) patients with brain metastasis (BM) and provide more references for predicting peritumoral edema. METHODS: We retrospectively reviewed 125 LUAD-BM patients who had undergone surgical resection from December 2015 to December 2020. The clinical characteristic, demographic, MRI data, and preNLR within 24-48 h before craniotomy were collected. Patients were divided into two groups based on preNLR (high NLR and low NLR), with cutoff values determined by receiver operating characteristic (ROC) analysis. Association between preoperative NLR and clinical features was determined by using Pearson chi-squared tests. Uni- and multivariate analyzes were performed to compare the overall survival (OS) of clinical features. RESULTS: The patients were divided into NLR-low (64 patients) and NLR-high (61 patients) groups based on receiver operating characteristic analysis of NLR area. According to correlation analysis, a high preNLR (NLR≥2.8) is associated with the both supra- and infratentorial location involved (P = 0.017) and a greater incidence of severe peritumoral edema (P = 0.038). By multivariable analysis, age ≥ 65 years (P = 0.011), KPS < 70 (P = 0.043), elevated preNLR (P = 0.013), extracerebral metastases (P = 0.003), EGFR/ALK+ (P = 0.037), postoperative radiotherapy (P = 0.017) and targeted therapy (P = 0.007) were independent prognostic factors. OS nomogram was constructed based on cox model and model performance was examined (AUC = 0.935). CONCLUSIONS: PreNLR may serve as a prognosis indicator in LUAD patients with brain metastasis, and high preNLR tends to be positively associate with multiple locations and severe peritumoral edema.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA