Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
World J Cardiol ; 16(8): 458-468, 2024 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-39221191

RESUMO

BACKGROUND: Cardio-oncology has received increasing attention especially among older patients with colorectal cancer (CRC). Cardiovascular disease (CVD)-specific mortality is the second-most frequent cause of death. The risk factors for CVD-specific mortality among older patients with CRC are still poorly understood. AIM: To identify the prognostic factors and construct a nomogram-based model to predict the CVD-specific mortality among older patients with CRC. METHODS: The data on older patients diagnosed with CRC were retrieved from The Surveillance, Epidemiology, and End Results database from 2004 to 2015. The prognostic factors and a nomogram-based model predicting the CVD-specific mortality were assessed using least absolute shrinkage and selection operator and Cox regression. RESULTS: A total of 141251 eligible patients with CRC were enrolled, of which 41459 patients died of CRC and 12651 patients died of CVD. The age at diagnosis, sex, marital status, year of diagnosis, surgery, and chemotherapy were independent prognostic factors associated with CVD-specific mortality among older patients with CRC. We used these variables to develop a model to predict CVD-specific mortality. The calibration curves for CVD-specific mortality probabilities showed that the model was in good agreement with actual observations. The C-index value of the model in the training cohort and testing cohort for predicting CVD-specific mortality was 0.728 and 0.734, respectively. CONCLUSION: The proposed nomogram-based model for CVD-specific mortality can be used for accurate prognostic prediction among older patients with CRC. This model is a potentially useful tool for clinicians to identify high-risk patients and develop personalized treatment plans.

2.
Math Biosci Eng ; 21(2): 2407-2431, 2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38454689

RESUMO

BACKGROUND: Aggrephagy is a lysosome-dependent process that degrades misfolded protein condensates to maintain cancer cell homeostasis. Despite its importance in cellular protein quality control, the role of aggrephagy in glioma remains poorly understood. OBJECTIVE: To investigate the expression of aggrephagy-related genes (ARGs) in glioma and in different cell types of gliomas and to develop an ARGs-based prognostic signature to predict the prognosis, tumor microenvironment, and immunotherapy response of gliomas. METHODS: ARGs were identified by searching the Reactome database. We developed the ARGs-based prognostic signature (ARPS) using data from the Cancer Genome Atlas (TCGA, n = 669) by Lasso-Cox regression. We validated the robustness of the signature in clinical subgroups and CGGA cohorts (n = 970). Gene set enrichment analysis (GSEA) was used to identify the pathways enriched in ARPS subgroups. The correlations between ARGs and macrophages were also investigated at single cell level. RESULTS: A total of 44 ARGs showed heterogeneous expression among different cell types of gliomas. Five ARGs (HSF1, DYNC1H1, DYNLL2, TUBB6, TUBA1C) were identified to develop ARPS, an independent prognostic factor. GSEA showed gene sets of patients with high-ARPS were mostly enriched in cell cycle, DNA replication, and immune-related pathways. High-ARPS subgroup had higher immune cell infiltration states, particularly macrophages, Treg cells, and neutrophils. APRS had positive association with tumor mutation burden (TMB) and immunotherapy response predictors. At the single cell level, we found ARGs correlated with macrophage development and identified ARGs-mediated macrophage subtypes with distinct communication characteristics with tumor cells. VIM+ macrophages were identified as pro-inflammatory and had higher interactions with malignant cells. CONCLUSION: We identified a novel signature based on ARGs for predicting glioma prognosis, tumor microenvironment, and immunotherapy response. We highlight the ARGs-mediated macrophages in glioma exhibit classical features.


Assuntos
Glioma , Macrófagos Associados a Tumor , Humanos , Macroautofagia , Sequência de Bases , Glioma/genética , Análise de Sequência de RNA , Microambiente Tumoral
3.
Free Radic Biol Med ; 209(Pt 2): 342-354, 2023 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-37898386

RESUMO

OBJECTIVES: N6-methyladenosine (m6A) is the most prevalent post-translational modification in eukaryotic mRNA. Recently, m6A editing modified by methyltransferase-like enzyme 3 (METTL3), the core m6A methyltransferase, has been demonstrated to be involved in cardiac sympathetic hyperactivity. This study aimed to clarify the effects and underlying mechanisms of METTL3 in the paraventricular nucleus (PVN) in mediating sympathetic activity following myocardial infarction (MI). METHODS: We established rat MI models by left anterior descending coronary artery ligation. m6A quantification was performed.The expression of METTL3 and its downstream gene, tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6), were determined. The functional role of METTL3 in sympathetic hyperactivity and electrical conduction stability were verified by assessing renal sympathetic nerve activity (RSNA), norepinephrine (NE) levels, and programmed electrical stimulation. Rescue experiments were also conducted. The mechanism by which m6A is involved in mitochondrial reactive oxygen species (mROS) production, mediated by TRAF6/ECSIT pathway, was explored in lipopolysaccharide (LPS) treated primary microglial cells. RESULTS: METTL3 was predominantly localized in the microglia and significantly increased within the PVN at 3 days post-MI. Inhibition of METTL3 decreased m6A levels, TRAF6 expression, and mROS production; downregulated sympathoexcitation, indicated by attenuated NE concentration and RSNA; decreased the incidence of ventricular tachycardia or fibrillation; and improved cardiac function. Mechanistically, downregulation of METTL3 prevented TRAF6 translocation to the mitochondria in the microglia and subsequent TRAF6/ECSIT pathway activation, resulting in decreased mROS production. CONCLUSIONS: This study demonstrates that METTL3-mediated m6A modification promotes sympathetic hyperactivity through TRAF6/ECSIT pathway and mitochondrial oxidative stress in the PVN, thereby leading to ventricular arrhythmias post-MI.


Assuntos
Infarto do Miocárdio , Fator 6 Associado a Receptor de TNF , Animais , Ratos , Metiltransferases/genética , Metiltransferases/metabolismo , Mitocôndrias/metabolismo , Infarto do Miocárdio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator 6 Associado a Receptor de TNF/genética , Fator 6 Associado a Receptor de TNF/metabolismo
4.
Rev Port Cardiol ; 42(2): 125-135, 2023 02.
Artigo em Inglês, Português | MEDLINE | ID: mdl-36759072

RESUMO

INTRODUCTION & OBJECTIVES: Acute myocardial infarction (AMI) in coronary heart disease is a leading cause of sudden death primarily due to malignant ventricular arrhythmias (VAs). Inflammatory cell infiltration and inflammation-induced overactivation of sympathetic nerves are the major cause of VAs in AMI pathophysiological processes. Type 2 macrophages play an anti-inflammatory role in AMI. Targeting macrophages may be a therapeutic strategy to prevent VAs post AMI. We found that gamma aminobutyric acid (GABA) promotes macrophages polarized to M2 and hypothesized that GABA might exert anti-inflammatory effects by promoting type 2 macrophage polarization in AMI. We aim to characterized GABAB receptor distribution, function, and mechanisms in M2 macrophage polarization and explored the functional aspect of GABAB receptor activation in sympathetic remodeling. RESULTS: Gamma aminobutyric acid B receptors were expressed on macrophage surface both in vitro and in vivo. GABAB receptor agonist baclofen, GABA promoted macrophage switch to M2. While GABAB receptor antagonist CGP52432 blocked a baclofen induced switch to M2 polarization. GABA and baclofen increased M2 macrophage percentage and CGP52432 blocked this process in vivo. Also, IL-10 and TGF-ß1 released by M2 were increased in both AMI and baclofen/AMI group; Serum NE levels were decreased by baclofen. All the above effects were reversed by CGP52432 treatment. Baclofen decreased TH and GAP-43 staining while CGP52432 enhanced their expression post AMI indicating GABAB receptor activation inhibited sympathetic nerve sprouting and activity by reducing NE release. CONCLUSIONS: Gamma aminobutyric acid B receptor activation promoted M2 polarization and protested AMI heart by regulating sympathetic nerve remodeling.


Assuntos
Infarto do Miocárdio , Receptores de GABA , Humanos , Baclofeno/farmacologia , Ácido gama-Aminobutírico/metabolismo , Macrófagos/metabolismo , Anti-Inflamatórios
5.
J Cardiol ; 79(3): 423-431, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34750029

RESUMO

BACKGROUND: Cardiac sympathetic hyperinnervation after myocardial infarction (MI) is associated with a high incidence of lethal arrhythmia. Erythropoietin-producing hepatoma interactor B2 (EphrinB2), a diffusible axonal chemorepellent that can induce growth cone collapse and axon repulsion of several neuronal populations, is crucial in neurodevelopment during disease development and progression. However, whether EphrinB2 could inhibit cardiac sympathetic hyperinnervation after MI remains unclear. METHODS AND RESULTS: A rat model of MI was developed by left anterior descending coronary artery ligation. EphrinB2 expression was markedly increased in the infarcted border at 3 days after MI. Downregulation of EphrinB2 by intramyocardial injection of lentivirus carrying EphrinB2-shRNA significantly increased sympathetic hyperinnervation along with downregulated RhoA expression. In contrast, injection of EphrinB2-overexpressing lentivirus markedly upregulated EphrinB2, concomitant with inhibition of sympathetic sprouting and upregulated RhoA expression, accompanied by decreased incidence of ventricular arrhythmias (VAs). However, co-administering EphrinB2-overexpressing lentivirus and Fasudil (Rho kinase inhibitor) nearly abolished the inhibition of nerve sprouting effect. Additionally, EphrinB2 expression did not affect nerve growth factor level in the infarcted heart. CONCLUSIONS: Overexpression of EphrinB2 may ameliorate MI-induced sympathetic hyperinnervation and further reduce the incidence of VAs, at least in part by activating RhoA-mediated axonal retraction.


Assuntos
Arritmias Cardíacas , Efrina-B2/metabolismo , Infarto do Miocárdio , Animais , Arritmias Cardíacas/complicações , Arritmias Cardíacas/prevenção & controle , Coração , Incidência , Infarto do Miocárdio/complicações , Miocárdio/metabolismo , Ratos , Sistema Nervoso Simpático , Regulação para Cima , Proteínas rho de Ligação ao GTP
6.
Redox Biol ; 24: 101186, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30978539

RESUMO

Sympathetic nerve hyperactivity is a primary reason for fatal ventricular arrhythmias (VAs) following myocardial infarction (MI). Pro-inflammatory cytokines produced in the paraventricular nucleus (PVN) post-MI are associated with sympathetic overexcitation; however, the precise mechanism needs further investigation. Our aim was to explore the mechanism of toll-like receptor 4 (TLR4) and its downstream molecular pathway in mediating sympathetic activity post-MI within the PVN. A rat MI model was developed via left anterior descending coronary artery ligation. TLR4 was primarily localized in microglia and increased markedly within the PVN at 3 days in MI rats. Sympathoexcitation also increased, as indicated by high levels of renal sympathetic nerve activity (RSNA) and norepinephrine (NE) concentration. TLR4 knockdown via shRNA microinjection to the PVN resulted in decreased activation of Fos protein (+) neurons in the PVN and peripheral sympathetic nerve activity. TLR4 knockdown also exhibited a lower arrhythmia score following programmed electrical stimulation than those treated with MI surgery only, indicating that the knockdown of TLR4 decreased the incidence of malignant ventricular arrhythmias following MI. LPS-induced inflammatory response was analyzed to explore the underlying mechanism of TLR4 in sympathetic hyperactivity. High levels of NF-κB protein, the pro-inflammatory cytokines IL-1ß and TNF-α, and ROS production were observed in the LPS group. PVN-targeted injection of the NF-κB inhibitor PDTC attenuated NF-κB expression and sympathetic activity. Taken together, the results suggested that knockdown of microglial TLR4 within the PVN decreased sympathetic hyperactivity and subsequent VAs post-MI. The downstream NF-κB pathway and ROS production participated in the process. Interventions targeting TLR4 signaling in the PVN may be a novel approach to ameliorate the incidence of VAs post-MI.


Assuntos
NF-kappa B/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Sistema Nervoso Simpático/metabolismo , Sistema Nervoso Simpático/fisiopatologia , Receptor 4 Toll-Like/metabolismo , Animais , Biomarcadores , Eletrocardiografia , Imunofluorescência , Imuno-Histoquímica , Masculino , Infarto do Miocárdio , Ratos
7.
J Cardiol ; 73(1): 81-88, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30487059

RESUMO

BACKGROUND: The hypothalamic paraventricular nucleus (PVN) is the center of the regulation of autonomic nervous system functions and cardiovascular activity. Phosphoinositide-3 kinase (PI3K)-AKT pathway in PVN contributes to mediate sympathetic nerve activity and is activated in spontaneously hypertensive rats. Overactivation of the sympathetic output was considered as an important mechanism of the arrhythmias. In the present study, we aimed to explore whether targeted regulation of sympathetic activity in PVN could reduce the peripheral sympathoexcitatory and attenuate the ventricular arrhythmias (VAs) in myocardial infarction (MI) rats via PI3K-AKT pathway. METHODS: A stainless steel gauge guide cannula was stereotaxically implanted into the PVN, and 7 days later, rats were randomly divided into the following 4 groups: group A, control+dimethyl sulfoxide (DMSO); group B, control+LY294002; group C, MI surgery+DMSO; and group D, MI surgery+LY294002. Studies were conducted seven days post-MI. Myocardial function, infarct size, inducible VAs by programmed electrical stimulation, renal sympathetic nerve activity (RSNA), and protein level of PI3K and AKT were measured. RESULTS: MI increased the protein ratios of p-PI3K-to-total-PI3K and p-AKT-to-total-AKT in PVN but can be reduced by LY294002 treatment. Inhibition of sympathetic nerve activity in PVN led to a reversion in plasma norepinephrine, RSNA and inducible VAs in MI rats. CONCLUSIONS: PI3K-AKT pathway in the PVN was a main mechanism in regulating sympathetic activity and arrhythmias in MI rats. Targeted inhibition of sympathetic activity in PVN may be a potential treatment for the VAs via PI3K-AKT pathway.


Assuntos
Arritmias Cardíacas/enzimologia , Infarto do Miocárdio/enzimologia , Núcleo Hipotalâmico Paraventricular/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Sistema Nervoso Simpático/enzimologia , Animais , Arritmias Cardíacas/fisiopatologia , Arritmias Cardíacas/prevenção & controle , Cromonas/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Masculino , Morfolinas/uso terapêutico , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/fisiopatologia , Miocárdio/enzimologia , Norepinefrina/sangue , Ratos , Ratos Endogâmicos SHR , Transdução de Sinais/efeitos dos fármacos
8.
J Cell Mol Med ; 23(1): 112-125, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30353660

RESUMO

Malignant ventricular arrhythmias (VAs) following myocardial infarction (MI) is a lethal complication resulting from sympathetic nerve hyperactivity. Numerous evidence have shown that inflammation within the paraventricular nucleus (PVN) participates in sympathetic hyperactivity. Our aim was to explore the role of Macrophage-inducible C-type lectin (Mincle) within the PVN in augmenting sympathetic activity following MI,and whether NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome/IL-1ß axis is involved in this activity. MI was induced by coronary artery ligation. Mincle expression localized in microglia within the PVN was markedly increased at 24 hours post-MI together with sympathetic hyperactivity, as indicated by measurement of the renal sympathetic nerve activity (RSNA) and norepinephrine (NE) concentration. Mincle-specific siRNA was administrated locally to the PVN, which consequently decreased microglial activation and sympathetic nerve activity. The MI rats exhibited a higher arrhythmia score after programmed electric stimulation than that treated with Mincle siRNA, suggesting that the inhibition of Mincle attenuated foetal ventricular arrhythmias post-MI. The underlying mechanism of Mincle in sympathetic hyperactivity was investigated in lipopolysaccharide (LPS)-primed naïve rats. Recombinant Sin3A-associated protein 130kD (rSAP130), an endogenous ligand for Mincle, induced high levels of NLRP3 and mature IL-1ß protein. PVN-targeted injection of NLRP3 siRNA or IL-1ß antagonist gevokizumab attenuated sympathetic hyperactivity. Together, the data indicated that the knockdown of Mincle in microglia within the PVN prevents VAs by attenuating sympathetic hyperactivity and ventricular susceptibility, in part by inhibiting its downstream NLRP3/IL-1ß axis following MI. Therapeutic interventions targeting Mincle signalling pathway could constitute a novel approach for preventing infarction injury.


Assuntos
Lectinas Tipo C/metabolismo , Microglia/metabolismo , Infarto do Miocárdio/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores Imunológicos/metabolismo , Sistema Nervoso Simpático/metabolismo , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Coração/fisiopatologia , Interleucina-1beta/antagonistas & inibidores , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lectinas Tipo C/genética , Masculino , Infarto do Miocárdio/genética , Infarto do Miocárdio/fisiopatologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Norepinefrina/metabolismo , Interferência de RNA , Ratos Sprague-Dawley , Receptores Imunológicos/genética , Sistema Nervoso Simpático/fisiopatologia
9.
Oncol Rep ; 34(6): 3097-103, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26398855

RESUMO

HSPC238 is a recently identified tumor suppressor and demonstrates ubiquitin ligase E3 enzyme activity. HSPC238 was found to be significantly downregulated in human hepatocellular carcinoma (HCC) in vivo and to inhibit the proliferation and invasion of hepatoma cells in vitro; however, the underlying molecular mechanism is largely unknown. In the present study, we screened for and identified proteins that physically interact with HSPC238. A bait vector for yeast two-hybrid was constructed with human HSPC238 gene cDNA. Yeast two-hybrid screening was performed using a human fetal liver cDNA library. Multiple reporter gene assays, DNA sequencing and BLAST comparison analysis were performed on positive clones. Protein interaction of screened candidates with HSPC238 was further validated by confocal microscopy, co-immunoprecipitation and pull-down assays. Yeast two-hybrid screening demonstrated 124 positive clones. Multiple reporter gene assays with LacZ, HIS and ADE2 selective media identified 12 genes. Further co-localization, co-immunoprecipitation and pull-down assays demonstrated that HMOX1, RPS27A, ubiquitinB and MT2A interacted with HSPC238. These four proteins are involved in tumor development and progression, and are associated with the ubiquitin-proteasome pathway. Our results suggest that HSPC238 may play a tumor suppressor role and interact with these proteins via the ubiquitin-proteasome pathway. The identification and validation of proteins interacting with HSP238 may lead to the discovery of novel mechanisms through which HSPC238 suppresses tumorigenesis in human hepatocellular carcinoma.


Assuntos
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Proteínas de Neoplasias/metabolismo , Ubiquitina-Proteína Ligases/genética , Carcinogênese/genética , Carcinoma Hepatocelular/patologia , Biblioteca Gênica , Vetores Genéticos , Humanos , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas/patologia , Proteínas de Neoplasias/genética , Ligação Proteica , Técnicas do Sistema de Duplo-Híbrido , Ubiquitina-Proteína Ligases/metabolismo
10.
Mol Cell Biochem ; 366(1-2): 131-7, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22584585

RESUMO

The gene for proteasome subunit alpha type-7 (PSMA7) is located in chromosomal 20q13.33, a region frequently amplified in tumor. In this study, we employed A549 human lung adenocarcinoma cells and showed that PSMA7 inhibits the proliferation, tumorigenicity and invasion of A549 cells in vitro. Moreover, both gain and loss of function studies demonstrated that PSMA7 modulates the tumorigenicity of A549 cells in a xenograft nude mice model. In conclusion, these results identify inhibitory effects associated with PSMA7 that affect the tumorigenicity of A549 cells, suggesting PSMA7 as a potential tumor biomarker.


Assuntos
Adenocarcinoma/metabolismo , Transformação Celular Neoplásica/metabolismo , Neoplasias Pulmonares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Transformação Celular Neoplásica/patologia , Feminino , Expressão Gênica , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/fisiologia , Interferência de RNA , Carga Tumoral
11.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 25(5): 402-5, 2009 May.
Artigo em Chinês | MEDLINE | ID: mdl-19426595

RESUMO

AIM: To construct the recombinant eukaryotic expression vector pDsRed1-C3/LOC51255 and investigate the subcellular localization of LOC51255 protein in HePG2 cell line. METHODS: The cDNA of LOC51255 was amplified from the recombinant plasmid pET28b/LOC51255 by PCR. The aim gene fragment LOC51255 from pMD18-T/LOC51255 was subcloned into eukaryotic expression vector pDsRed1-C3. The recombinant plasmid pDsRed1-C3/LOC51255 was identificated by BamH I/Xho I double digestion and sequence analysis, and then transfected into HePG2 cell line by lipofectamine 2000. The expression of LOC51255 protein and its subcellular localization were detected by fluorescence microscope. RESULTS: The construction of the recombinant plasmid pDsRed1-C3/LOC51255 was proved by restriction enzyme digestion analysis and DNA sequencing. Its red fluorescent protein was mainly detected in the cytoplasm of HePG2 cell line. CONCLUSION: The eukaryotic expression plasmid pDsRed1-C3/LOC51255 has been successfully constructed and expressed in the HepG2 cell line. LOC51255 protein has been proved to be located in the cytoplasm of HePG2 cell line. Our research will help further studies on the function of human gene LOC51255.


Assuntos
Citoplasma/metabolismo , Vetores Genéticos/genética , Proteínas Recombinantes de Fusão/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Linhagem Celular Tumoral , Clonagem Molecular , Células Eucarióticas/metabolismo , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microscopia de Fluorescência , Proteínas Recombinantes de Fusão/genética , Análise de Sequência de DNA , Transfecção , Ubiquitina-Proteína Ligases/genética , Proteína Vermelha Fluorescente
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA