Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 81(1): 167, 2024 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-38581570

RESUMO

Hepatocellular carcinoma (HCC) is a malignant tumor with high incidence and mortality rates. NFKBIZ, a member of the nuclear factor kappa B inhibitory family, is closely related to tumor progression. However, the precise role of NFKBIZ in HCC remains unclear. To explore this, we conducted a series of experiments from clinic to cells. Western blot and qPCR revealed a significant downregulation of NFKBIZ in human HCC tissues. Clinical character analysis showed that the patients with lower NFKBIZ expression had poorer prognosis and higher clinical stage. By using CCK-8, wound healing, transwell invasion and migration assay, we discovered that NFKBIZ expression was reversely associated with the proliferation, invasion, and migration ability of HCC cells in vitro. Additionally, the results obtained from xenograft assay and lung metastasis models showed that NFKBIZ overexpression inhibited the growth and metastasis of HCC cells in vivo. Western blot and immunofluorescence assay further revealed that NFKBIZ mediated HCC cell growth and migration by regulating NFκB signaling transduction. Finally, flow cytometry, protein degradation assay and Co-immunoprecipitation indicated that TRIM16 can enhance NFKBIZ ubiquitination by direct interactions at its K48 site, which may thereby alleviate HCC cell apoptosis to induce the insensitivity to sorafenib. In conclusion, our study demonstrated that NFKBIZ regulated HCC tumorigenesis and metastasis by mediating NFκB signal transduction and TRIM16/NFKBIZ/NFκB axis may be the underlying mechanism of sorafenib insensitivity in HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Sorafenibe/farmacologia , Linhagem Celular Tumoral , Movimento Celular , Transdução de Sinais , Carcinogênese/genética , Transformação Celular Neoplásica , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo
2.
Hepatology ; 75(2): 338-352, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34455616

RESUMO

BACKGROUND AND AIMS: Hepatic ischemia-reperfusion injury (IRI) is a common complication of hepatectomy and liver transplantation. However, the mechanisms underlying hepatic IRI have not been fully elucidated. Regulator of G-protein signaling 14 (RGS14) is a multifunctional scaffolding protein that integrates the G-protein and mitogen-activated protein kinase (MAPK) signaling pathways. However, the role of RGS14 in hepatic IRI remains unclear. APPROACH AND RESULTS: We found that RGS14 expression increased in mice subjected to hepatic ischemia-reperfusion (IR) surgery and during hypoxia reoxygenation in hepatocytes. We constructed global RGS14 knockout (RGS14-KO) and hepatocyte-specific RGS14 transgenic (RGS14-TG) mice to establish 70% hepatic IRI models. Histological hematoxylin and eosin staining, levels of alanine aminotransferase and aspartate aminotransferase, expression of inflammatory factors, and apoptosis were used to assess liver damage and function in these models. We found that RGS14 deficiency significantly aggravated IR-induced liver injury and activated hepatic inflammatory responses and apoptosis in vivo and in vitro. Conversely, RGS14 overexpression exerted the opposite effect of the RGS14-deficient models. Phosphorylation of TGF-ß-activated kinase 1 (TAK1) and its downstream effectors c-Jun N-terminal kinase (JNK) and p38 increased in the liver tissues of RGS14-KO mice but was repressed in those of RGS14-TG mice. Furthermore, inhibition of TAK1 phosphorylation rescued the effect of RGS14 deficiency on JNK and p38 activation, thus blocking the inflammatory responses and apoptosis. CONCLUSIONS: RGS14 plays a protective role in hepatic IR by inhibiting activation of the TAK1-JNK/p38 signaling pathway. This may be a potential therapeutic strategy for reducing incidences of hepatic IRI in the future.


Assuntos
MAP Quinase Quinase Quinases/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo , Alanina Transaminase/metabolismo , Animais , Apoptose , Aspartato Aminotransferases/metabolismo , Hipóxia Celular , Células Cultivadas , Ativação Enzimática , Hepatócitos/metabolismo , Inflamação/genética , Inflamação/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fígado/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fosforilação , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
Hepatology ; 76(1): 94-111, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-34735734

RESUMO

BACKGROUND AND AIMS: Hepatic ischemia-reperfusion (HIR) injury, a common clinical complication of liver transplantation and resection, affects patient prognosis. Ring finger protein 5 (RNF5) is an E3 ubiquitin ligase that plays important roles in endoplasmic reticulum stress, unfolded protein reactions, and inflammatory responses; however, its role in HIR is unclear. APPROACH AND RESULTS: RNF5 expression was significantly down-regulated during HIR in mice and hepatocytes. Subsequently, RNF5 knockdown and overexpression of cell lines were subjected to hypoxia-reoxygenation challenge. Results showed that RNF5 knockdown significantly increased hepatocyte inflammation and apoptosis, whereas RNF5 overexpression had the opposite effect. Furthermore, hepatocyte-specific RNF5 knockout and transgenic mice were established and subjected to HIR, and RNF5 deficiency markedly aggravated liver damage and cell apoptosis and activated hepatic inflammatory responses, whereas hepatic RNF5 transgenic mice had the opposite effect compared with RNF5 knockout mice. Mechanistically, RNF5 interacted with phosphoglycerate mutase family member 5 (PGAM5) and mediated the degradation of PGAM5 through K48-linked ubiquitination, thereby inhibiting the activation of apoptosis-regulating kinase 1 (ASK1) and its downstream c-Jun N-terminal kinase (JNK)/p38. This eventually suppresses the inflammatory response and cell apoptosis in HIR. CONCLUSIONS: We revealed that RNF5 protected against HIR through its interaction with PGAM5 to inhibit the activation of ASK1 and the downstream JNK/p38 signaling cascade. Our findings indicate that the RNF5-PGAM5 axis may be a promising therapeutic target for HIR.


Assuntos
Proteínas de Membrana , Fosfoproteínas Fosfatases , Traumatismo por Reperfusão , Ubiquitina-Proteína Ligases , Animais , Apoptose , Humanos , Fígado/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Fosfoproteínas Fosfatases/metabolismo , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
4.
J Enzyme Inhib Med Chem ; 38(1): 2237701, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37489043

RESUMO

In this work, a series of novel arylamide derivatives containing piperazine moiety were designed and synthesised as tubulin polymerisation inhibitors. Among 25 target compounds, compound 16f (MY-1121) exhibited low nanomolar IC50 values ranging from 0.089 to 0.238 µM against nine human cancer cells. Its inhibitory effects on liver cancer cells were particularly evident with IC50 values of 89.42 and 91.62 nM for SMMC-7721 and HuH-7 cells, respectively. Further mechanism studies demonstrated that compound 16f (MY-1121) could bind to the colchicine binding site of ß-tubulin and directly act on ß-tubulin, thus inhibiting tubulin polymerisation. Additionally, compound 16f (MY-1121) could inhibit colony forming ability, cause morphological changes, block cell cycle arrest at the G2 phase, induce cell apoptosis, and regulate the expression of cell cycle and cell apoptosis related proteins in liver cancer cells. Overall, the promising bioactivities of compound 16f (MY-1121) make the novel arylamide derivatives have the value for further development as tubulin polymerisation inhibitors with potent anticancer activities.


Assuntos
Neoplasias Hepáticas , Tubulina (Proteína) , Humanos , Apoptose , Sítios de Ligação , Piperazina , Moduladores de Tubulina
5.
Drug Dev Res ; 84(2): 211-225, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36567664

RESUMO

Acetaminophen (APAP) overdose-induced acute liver injury (ALI) causes hepatocyte cell death, oxidative stress, and inflammation. Oridonin (Ori), a covalent NLRP3-inflammasome inhibitor, ameliorates APAP-induced ALI through an unclear molecular mechanism. This study found that Ori decreased hepatic cytochrome P450 2E1 level and increased glutathione content to prevent APAP metabolism, and then reduced the necrotic area, improved liver function, and inhibited APAP-induced proinflammatory cytokines and oxidative stress. Ori also decreased activating transcription factor 4 (ATF4) protein levels and increased peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) to reduce APAP-induced endoplasmic reticulum stress activation and mitochondrial dysfunction. Furthermore, western blot and luciferase assay found that ATF4 inhibited transcription in the PGC-1α promoter -507 to -495 region to reduce PGC-1α levels, while ATF4 knockdown neutralized the hepatoprotective effect of Ori. Molecular docking showed that Ori bound to ATF4's amino acid residue glutamate 302 through 6, 7, and 18 hydroxyl bands. Our findings demonstrated that Ori prevented metabolic activation of APAP and further inhibited the ATF4/PGC-1α pathway to alleviate APAP overdose-induced hepatic toxicity, which illuminated its potential therapeutic effects on ALI.


Assuntos
Acetaminofen , Doença Hepática Induzida por Substâncias e Drogas , Humanos , Acetaminofen/efeitos adversos , Fator 4 Ativador da Transcrição/metabolismo , Fator 4 Ativador da Transcrição/farmacologia , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Fígado , Simulação de Acoplamento Molecular , Estresse Oxidativo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo
6.
Mol Biol Rep ; 48(2): 1687-1695, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33484391

RESUMO

Ischemia-reperfusion injury (IRI) is inevitable during liver surgery, and it is an important factor affecting the prognosis of patients. IL-6 rs1800796 single nucleotide polymorphisms (SNPs) can promote synthesis and secretion of IL-6 and protect hepatocytes from IRI. In this study, we investigated the mechanisms by which IL-6 alleviates hepatic IRI. We transfected lentivirus which carries IL-6 rs1800796 to L02 cells and constructed the cell line (L02-IL6) with a high expression of IL-6. The biological function of IL-6 SNPs was explored through a cell model of hypoxia-reoxygenation (H/R). Cell viability was evaluated by CCK8 and Real-Time Cell Analysis (RTCA), and found that the viability of the L02-IL6 cells was higher than that of the control group (P < 0.01). Flow cytometry assay showed that the rate of apoptosis was significantly decreased in L02-IL6 cells. Furthermore, in comparison with the control group, the level of cleaved-caspase3, which is an important marker of apoptosis, was dramatically decreased. These differences showed that the sequence variants at rs1800796 of the IL-6 gene could improve the resistance against H/R. Moreover, the levels of autophagy-related proteins, such as LC3 and Beclin-1, were upregulated in L02-IL6 group on H/R injury, which means IL-6 could alleviate apoptosis via activating the autophagy pathway. And we also found that the STAT3 signal pathway was activated. Next, we investigated whether the exogenous treatment with IL-6 affect hepatocytes and thus play a protective role. We pre-treated the L02 cells with recombinant human IL-6 for 12 h and then made H/R treatment. We found the treatment with 100 ng/ml IL-6 alleviated the damage of L02 cells and inhibited the apoptosis. And the further study revealed the pre-treatment with IL-6 activated the STAT3 signaling pathway in the L02 cells and then caused the activation of autophagy and apoptosis inhibition. IL-6 might play a critical role in alleviating hepatic IRI, through its modulation of the STAT3 signaling pathway, and activation of autophagy. Recombinant human IL-6 might be a potential therapeutic target in hepatic IRI.


Assuntos
Apoptose/genética , Autofagia/genética , Hepatócitos/metabolismo , Interleucina-6/genética , Interleucina-6/farmacologia , Fígado/efeitos dos fármacos , Traumatismo por Reperfusão/metabolismo , Fator de Transcrição STAT3/metabolismo , Autofagia/efeitos dos fármacos , Caspase 3/genética , Caspase 3/metabolismo , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/genética , Linhagem Celular , Sobrevivência Celular/genética , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Humanos , Interleucina-6/metabolismo , Fígado/metabolismo , Fígado/patologia , Polimorfismo de Nucleotídeo Único , Proteínas Recombinantes , Traumatismo por Reperfusão/genética , Fator de Transcrição STAT3/genética , Transdução de Sinais/genética , Regulação para Cima
7.
Eur Radiol ; 29(11): 6191-6201, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31041565

RESUMO

OBJECTIVES: To evaluate the performance of a novel three-dimensional (3D) joint convolutional and recurrent neural network (CNN-RNN) for the detection of intracranial hemorrhage (ICH) and its five subtypes (cerebral parenchymal, intraventricular, subdural, epidural, and subarachnoid) in non-contrast head CT. METHODS: A total of 2836 subjects (ICH/normal, 1836/1000) from three institutions were included in this ethically approved retrospective study, with a total of 76,621 slices from non-contrast head CT scans. ICH and its five subtypes were annotated by three independent experienced radiologists, with majority voting as reference standard for both the subject level and the slice level. Ninety percent of data was used for training and validation, and the rest 10% for final evaluation. A joint CNN-RNN classification framework was proposed, with the flexibility to train when subject-level or slice-level labels are available. The predictions were compared with the interpretations from three junior radiology trainees and an additional senior radiologist. RESULTS: It took our algorithm less than 30 s on average to process a 3D CT scan. For the two-type classification task (predicting bleeding or not), our algorithm achieved excellent values (≥ 0.98) across all reporting metrics on the subject level. For the five-type classification task (predicting five subtypes), our algorithm achieved > 0.8 AUC across all subtypes. The performance of our algorithm was generally superior to the average performance of the junior radiology trainees for both two-type and five-type classification tasks. CONCLUSIONS: The proposed method was able to accurately detect ICH and its subtypes with fast speed, suggesting its potential for assisting radiologists and physicians in their clinical diagnosis workflow. KEY POINTS: • A 3D joint CNN-RNN deep learning framework was developed for ICH detection and subtype classification, which has the flexibility to train with either subject-level labels or slice-level labels. • This deep learning framework is fast and accurate at detecting ICH and its subtypes. • The performance of the automated algorithm was superior to the average performance of three junior radiology trainees in this work, suggesting its potential to reduce initial misinterpretations.


Assuntos
Algoritmos , Aprendizado Profundo , Imageamento Tridimensional/métodos , Hemorragias Intracranianas/diagnóstico , Tomografia Computadorizada por Raios X/métodos , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Reprodutibilidade dos Testes , Estudos Retrospectivos , Adulto Jovem
8.
Immunology ; 154(1): 132-143, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29197065

RESUMO

Exosomes derived from heat-stressed tumour cells (HS-TEXs), which contain abundant heat shock protein (HSP) 70, strongly induce antitumour immune responses. HSP70-induced interleukin (IL)-6 promotes IL-17 expression and causes rejection of established prostate tumours. However, it remains unclear whether HS-TEXs exhibit antitumour effects by converting regulatory T cells (Tregs ) into T helper type 17 (Th17) cells. In this study, we found that compared with TEXs, HS-TEXs were more potent in stimulating secretion of IL-6 from dendritic cells. In vitro, IL-6 blocked tumour cell-derived transforming growth factor beta 1-induced Treg differentiation and promoted Th17 cell differentiation. HS-TEXs exerted strong antitumour effects, converting Tregs into Th17 cells with high efficiency, a process that was entirely dependent upon IL-6. Neutralization of IL-17 completely abolished the antitumour effect of TEXs, but only partially inhibited that of HS-TEXs. In addition, we found higher levels of IL-6 and IL-17 in serum from tumour patients treated with hyperthermia, and an increase in Th17 cells and a decrease in Tregs was detected in peripheral blood mononuclear cells isolated from these patients after hyperthermia. Therefore, our results demonstrate that HS-TEXs possess a powerful capacity to convert immunosuppressive Tregs into Th17 cells via IL-6, which contributes to their potent antitumour effect.


Assuntos
Adenocarcinoma/terapia , Proliferação de Células , Neoplasias do Colo/terapia , Exossomos/transplante , Hipertermia Induzida/métodos , Interleucina-6/imunologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Adenocarcinoma/imunologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adulto , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Exossomos/imunologia , Exossomos/metabolismo , Exossomos/patologia , Feminino , Resposta ao Choque Térmico , Humanos , Interleucina-6/sangue , Interleucina-6/metabolismo , Linfócitos do Interstício Tumoral/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Transdução de Sinais , Linfócitos T Reguladores/metabolismo , Células Th17/metabolismo , Fatores de Tempo , Carga Tumoral , Microambiente Tumoral
9.
Cell Signal ; 114: 110994, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38036196

RESUMO

Primary liver cancer is known for its high incidence and fatality rate. Over the years, therapeutic strategies for primary liver cancer have advanced significantly. Nonetheless, a substantial number of patients have not benefited from these methods, underscoring the pressing need for new and effective treatments for primary liver cancer. Ubiquitination is a critical post-translational modification that enables proteins to fulfill their normal biological functions and maintain their expression stability within cells. Importantly, increasing evidence suggests that the progression of liver cancer cells is often accompanied by disruptions in protein ubiquitination and deubiquitination processes. In this comprehensive review, we have compiled pertinent research about dysregulated ubiquitination in hepatocellular carcinoma (HCC) to broaden our understanding in this field. We elucidate the connections between the ubiquitination proteasome system, deubiquitination, and HCC. Furthermore, we shed light on the role of ubiquitination in cells situated within the tumor microenvironment of HCC including its involvement in mediating the activation of oncogenic pathways, reprogramming metabolic processes, and perturbing normal cellular functions. In conclusion, targeting the dysregulation of ubiquitination in HCC holds promise as a prospective and complementary therapeutic approach to existing treatments.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Estudos Prospectivos , Ubiquitinação , Processamento de Proteína Pós-Traducional , Microambiente Tumoral
10.
J Cancer Res Clin Oncol ; 150(6): 303, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38861187

RESUMO

BACKGROUND: Human MARCH5 is a mitochondria-localized E3 ubiquitin-protein ligase that is essential for the regulation of mitochondrial dynamics. A large body of evidence suggests that imbalances in mitochondrial dynamics are strongly associated with cancer. However, the expression, biological function and prognostic significance of MARCH5 in hepatocellular carcinoma (HCC) have not been determined. MATERIALS AND METHODS: The mRNA and protein expression of MARCH5 in HCC cell lines and tumor tissues was assessed by real-time quantitative PCR, Western blot analysis and immunohistochemistry. The clinical prognostic significance of MARCH5 was evaluated in 135 HCC patients. Knockdown or overexpression of MARCH5 in HCC cells was determined by in vitro cell proliferation, migration and invasion assays, and in vivo tumor growth and metastasis assays. In addition, the intrinsic mechanisms by which MARCH5 regulates HCC cell growth and metastasis were explored. RESULTS: MARCH5 was significantly overexpressed in HCC cells and was closely associated with patients' poor postoperative prognosis. In vivo and in vitro experiments revealed that MARCH5 significantly promoted the increase and invasive and migratory ability of hepatocellular carcinoma cells, which was mainly due to the promotion of autophagy by MARCH5. Mechanistic studies revealed that MARCH5 promoted autophagy through ubiquitination degradation of p53 leading to malignant progression of hepatocellular carcinoma. CONCLUSION: Our findings suggest that MARCH5 plays a critical oncogenic role in HCC cells, which provides experimental evidence for the use of MARCH5 as a potential target for HCC therapy.


Assuntos
Carcinoma Hepatocelular , Progressão da Doença , Neoplasias Hepáticas , Camundongos Nus , Proteína Supressora de Tumor p53 , Ubiquitina-Proteína Ligases , Ubiquitinação , Humanos , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/genética , Masculino , Animais , Camundongos , Feminino , Prognóstico , Proliferação de Células , Pessoa de Meia-Idade , Linhagem Celular Tumoral , Camundongos Endogâmicos BALB C , Movimento Celular
11.
Sci Rep ; 14(1): 8535, 2024 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-38609411

RESUMO

Although the death of hepatocytes is a crucial trigger of liver ischemia-reperfusion (I/R) injury, the regulation of liver I/R-induced hepatocyte death is still poorly understood. Phosphoglycerate mutase 5 (PGAM5), a mitochondrial Serine/Threonine protein phosphatase, regulates mitochondrial dynamics and is involved in the process of both apoptosis and necrotic. However, it is still unclear what role PGAM5 plays in the death of hepatocytes induced by I/R. Using a PGAM5-silence mice model, we investigated the role of PGAM5 in liver I/R injury and its relevant molecular mechanisms. Our data showed that PGAM5 was highly expressed in mice with liver I/R injury. Silence of PGAM5 could decrease I/R-induced hepatocyte death in mice. In subcellular levels, the silence of PGAM5 could restore mitochondrial membrane potential, increase mitochondrial DNA copy number and transcription levels, inhibit ROS generation, and prevent I/R-induced opening of abnormal mPTP. As for the molecular mechanisms, we indicated that the silence of PGAM5 could inhibit Drp1(S616) phosphorylation, leading to a partial reduction of mitochondrial fission. In addition, Mdivi-1 could inhibit mitochondrial fission, decrease hepatocyte death, and attenuate liver I/R injury in mice. In conclusion, our data reveal the molecular mechanism of PGAM5 in driving hepatocyte death through activating mitochondrial fission in liver I/R injury.


Assuntos
Fosfoglicerato Mutase , Traumatismo por Reperfusão , Animais , Camundongos , Hepatócitos , Fígado , Dinâmica Mitocondrial , Fosfoglicerato Mutase/genética , Traumatismo por Reperfusão/genética
12.
Psychol Res Behav Manag ; 17: 1831-1840, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38707965

RESUMO

Purpose: This study aims to translate and validate the Learned Helplessness Scale (LHS) for use in the educational context and specifically among Chinese law school students. Understanding learned helplessness in the context of Chinese law students can provide unique insights into the interaction of legal education, psychological health, and cultural influences, thereby contributing to a more nuanced understanding of learned helplessness. Methods: A total of 711 Chinese college students from two law schools participated in this study. The Learned Helplessness Scale (LHS) was translated into Chinese using forward and backward translation. Exploratory and confirmatory factor analysis, and construct validity were conducted to assess the dimensionality of the Chinese version of the LHS (Chinese LHS). Results: The exploratory factor analysis indicated that the Chinese LHS has a four-factor structure consisting of 14 items, which accounted for 50% of the total variance. The subsequent confirmatory factor analysis further supported this four-factor structure. The internal consistency of the Chinese LHS was found to be medium to high, with Cronbach's α values ranging from 0.63 to 0.87 for the subfactors, and 0.79 for the total scale. In addition, concurrent validity is also confirmed. Conclusion: The 14-item version of the Chinese LHS is a psychometric sound instrument for assessing learned helplessness among Chinese law school students.

13.
Tumour Biol ; 34(4): 2403-7, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23625656

RESUMO

Glutathione S-transferases (GSTs) play important roles in the detoxification of electrophilic carcinogens, and GSTM1 null genotype is associated with the dysfunction of GSTs. Previous studies investigating the association between GSTM1 null genotype and risk of esophageal carcinoma in Chinese provided inconsistent findings. To provide a more precise estimation on the association between GSTM1 null genotype and risk of esophageal carcinoma in Chinese population, a meta-analysis was performed. Eligible studies were searched in PubMed, Embase, and China National Knowledge Infrastructure databases. Odds ratio (OR) with the corresponding 95 % confidence interval (95 %CI) was used to assess the association. A total of 18 case-control studies involving 1,947 cases and 3,506 controls were finally included in the meta-analysis. Meta-analysis of those 18 studies showed that GSTM1 null genotype was associated with an increased risk of esophageal carcinoma in Chinese (random effect model OR = 1.49, 95 %CI = 1.11-2.00, P = 0.008). The findings from cumulative meta-analysis showed that the association was more obvious as the data increased by publication year. There was no risk of publication bias in the meta-analysis. Therefore, the findings from our meta-analysis provide a strong evidence for the association between GSTM1 null genotype and risk of esophageal carcinoma in Chinese population, and GSTM1 null genotype contributes to increased risk of esophageal carcinoma in Chinese.


Assuntos
Neoplasias Esofágicas/genética , Glutationa Transferase/genética , Povo Asiático/genética , Estudos de Casos e Controles , China , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Razão de Chances , Risco
14.
Food Chem Toxicol ; 181: 114050, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37734463

RESUMO

Doxorubicin induced cardiotoxicity (DIC) arises from mitochondrial dysfunction and oxidative stress. Oridonin (Ori), a natural tetracycline diterpenoid, has shown cardiac protective effect; however, its role in DIC remains unclear. This study investigates the protective effect of Ori against DIC and elucidates its underlying molecular mechanisms. The results demonstrate that Ori significantly alleviated DIC by improving myocardial structure, reducing the proportion of apoptotic cells, and alleviating the myocardial oxidative damage and mitochondrial dysfunction both in vivo and in vitro. Doxorubicin significantly decreased Sirt6 and PGC1α levels in cardiac tissues, which was reversed by Ori. Furthermore, Sirt6 overexpression significantly improved myocardial structure and reduced the proportion of apoptotic cells by reducing oxidative stress and improving mitochondrial function. The protective effect of Ori is neutralized by the Sirt6 inhibitor OSS_128167, evidenced by downregulated mRNA and protein expression of PGC1α. The transcription factor E2F1 was upregulated by doxorubicin, leading to decreased Sirt6 expression-an effect mitigated by Ori. Molecular docking simulations indicate direct binding between Ori and specific amino acid residues on E2F1 through hydroxyl bonds. These findings uncover a novel mechanism whereby Ori attenuates DIC by modulating the E2F1/Sirt6/PGC1α pathway.


Assuntos
Doxorrubicina , Sirtuínas , Camundongos , Animais , Doxorrubicina/toxicidade , Transdução de Sinais , Cardiotoxicidade/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Simulação de Acoplamento Molecular , Estresse Oxidativo , Miócitos Cardíacos , Sirtuínas/genética , Sirtuínas/metabolismo , Sirtuínas/farmacologia
15.
Cell Rep ; 42(1): 112002, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36662617

RESUMO

Type I interferon (IFN-I) signaling is central to inducing antiviral innate immunity. However, the mechanisms for IFN-I signaling self-regulation are still largely unknown. Here, we report that RNA virus-infected macrophages with UBE2M deficiency produced decreased IFN-I expression in a RIG-I-dependent manner, causing an aggravated viral infection. Mechanistically, UBE2M inhibits RIG-I degradation by preventing the interaction of RIG-I and E3 ligase STUB1, resulting in antiviral IFN-I signaling activation. Simultaneously, IFN-I signaling-activated STAT1 facilitates the transcription of Trim21, leading to increased UBE2M degradation and blunted antiviral immunity. Translationally, oral administration of milk-derived extracellular vesicles containing RING domain-truncated TRIM21 (TRIM21-ΔRING) lacking E3 ligase activity efficiently transfers TRIM21-ΔRING into macrophages. TRIM21-ΔRING suppresses UBE2M degradation by competitively binding to UBE2M with TRIM21, thereby enhancing antiviral immunity. Overall, we reveal a negative feedback loop of IFN-I signaling and develop a reagent to improve innate immunity against RNA viruses.


Assuntos
Interferon Tipo I , Vírus de RNA , Antivirais , Retroalimentação , Imunidade Inata , Ubiquitina-Proteína Ligases/genética
16.
Front Immunol ; 13: 937406, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36131916

RESUMO

The tumor microenvironment (TME) has become a major research focus in recent years. The TME differs from the normal extracellular environment in parameters such as nutrient supply, pH value, oxygen content, and metabolite abundance. Such changes may promote the initiation, growth, invasion, and metastasis of tumor cells, in addition to causing the malfunction of tumor-infiltrating immunocytes. As the neoplasm develops and nutrients become scarce, tumor cells transform their metabolic patterns by reprogramming glucose, lipid, and amino acid metabolism in response to various environmental stressors. Research on carcinoma metabolism reprogramming suggests that like tumor cells, immunocytes also switch their metabolic pathways, named "immunometabolism", a phenomenon that has drawn increasing attention in the academic community. In this review, we focus on the recent progress in the study of lipid metabolism reprogramming in immunocytes within the TME and highlight the potential target molecules, pathways, and genes implicated. In addition, we discuss hypoxia, one of the vital altered components of the TME that partially contribute to the initiation of abnormal lipid metabolism in immune cells. Finally, we present the current immunotherapies that orchestrate a potent antitumor immune response by mediating the lipid metabolism of immunocytes, highlight the lipid metabolism reprogramming capacity of various immunocytes in the TME, and propose promising new strategies for use in cancer therapy.


Assuntos
Neoplasias , Microambiente Tumoral , Aminoácidos/metabolismo , Glucose , Humanos , Metabolismo dos Lipídeos , Lipídeos , Neoplasias/patologia , Oxigênio
17.
Front Public Health ; 10: 1015446, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36523580

RESUMO

Uncertainty is mushrooming throughout COVID-19, and intolerance of uncertainty (IoU) nudges people into mental health difficulties involving fear, depression, and anxiety. The objective of this study was to investigate the role of depression and fear of COVID-19 (FoC) in the association between IoU and future career anxiety (FCA) among Chinese university students during the COVID-19 pandemic. This study involved 1,919 Chinese undergraduate students from 11 universities in eight Chinese cities with an online self-administered survey that included demographic information, IoU, FoC, depression, and FCA completed by all participants. Our study demonstrated a positive relationship between IoU and FCA and the chain mediation effect of FoC and depression. Thus, understanding how FoC affects FCA not only informs university career professionals and assists students in preparing for employment, but also motivates schools to offer career opportunities workshops and, most importantly, provides mental health support to help students effectively cope with uncertainty and overcome COVID-19-related stress.


Assuntos
COVID-19 , Humanos , COVID-19/epidemiologia , Pandemias , Incerteza , Depressão/epidemiologia , Depressão/psicologia , População do Leste Asiático , Ansiedade/epidemiologia , Ansiedade/psicologia , Estudantes/psicologia , Medo
18.
Int Immunopharmacol ; 113(Pt B): 109469, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36435062

RESUMO

Timely treatment of acute inflammatory reactions induced by fungi or bacteria is essential to prevent infectious damage. Ibrutinib is a Bruton's tyrosine kinase (BTK) inhibitor which is used to treat various lymphoid cancers. It is also known that BTK plays important roles in innate immunity and inflammatory response. In the present study, we investigated the regulatory effects of Ibrutinib on the activation of neutrophils and macrophages and its therapeutic effects on acute peritonitis. In addition, we also studied its anti-inflammatory mechanisms. The results showed that Ibrutinib inhibited the expression and secretion of inflammatory factors in macrophages induced by multiple Toll-like receptor (TLR) agonists. In the study of neutrophils, Ibrutinib selectively suppressed the activation, superoxide release, and calcium influx of neutrophils stimulated by zymosan. Furthermore, in zymosan-induced mice acute peritonitis, Ibrutinib significantly reduced the infiltration of neutrophils into peritoneal cavity, the release of myeloperoxidase (MPO) and ß-glucuronidase as well as the production of inflammatory factors in peritoneal cavity. In mechanism study, Ibrutinib selectively inhibited the phosphorylation of PLCγ2, PKCδ, and ERK1/2 in neutrophils induced by zymosan. Collectively, Ibrutinib can significantly inhibit the activation of neutrophils and macrophages by inhibiting BTK-PLCγ2-PKC signaling pathway, and has great potential to be developed into therapeutic drug for acute inflammatory diseases.


Assuntos
Neutrófilos , Peritonite , Animais , Camundongos , Zimosan , Fosfolipase C gama , Macrófagos , Adjuvantes Imunológicos , Peritonite/induzido quimicamente , Peritonite/tratamento farmacológico
19.
J Environ Pathol Toxicol Oncol ; 41(4): 11-23, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36374959

RESUMO

Gestational diabetes mellitus (GDM), a common complication in pregnancy, could threaten the health of both pregnancies and their offspring. miR-210-3p has been reported that play a crucial role in many diseases. Nevertheless, the molecular mechanism and clinical significance of miR-210-3p in the GDM is still unclear. miR-210-3p was overexpressed in the pancreas of the GDM mouse model. Meanwhile, miR-210-3p weakens cell viability and promotes the apoptosis of pancreatic ß cells, impairing the function of pancreatic ß cells. Bioinformatics analysis showed that miR-210-3p directly targets the expression of Dtx1, and miR-210-3p negatively regulated dtx1. Down-expression of Dtx1 could increase the expression of insulin and boost the function of pancreatic ß cells through inhibiting expressions of p-Akt, p-mTOR, p-4E-BP1, and p-SGK1. Rescue experiments verified that miR-210-3p could regulate the function of pancreatic ß cells and adjust the content of TG, TC, and HDL in the blood of mice with GDM via regulating the expression of Dtx1. The study demonstrated that miR-210-3p is significantly overexpressed in the pancreas of the GDM mouse model, which could impair the function and cell viability of pancreatic ß cells via suppressing the expression of Dtx1 promotes the progression of GDM. These findings provide a novel strategy to treat GDM.


Assuntos
Diabetes Gestacional , Células Secretoras de Insulina , MicroRNAs , Ubiquitina-Proteína Ligases , Animais , Feminino , Humanos , Camundongos , Gravidez , Apoptose , Diabetes Gestacional/genética , Insulina , Células Secretoras de Insulina/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
20.
Biomed Pharmacother ; 155: 113635, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36095957

RESUMO

Non-SMC Condensin I complex subunit G (NCAPG), a mitosis-associated chromosomal condensation protein, is related to sister chromatid appropriate separation during the condensation and fusion of chromosomes and responsible for the condensation and stabilization of chromosomes during meiosis and mitosis. Studies have shown that NCAPG is highly adjusted in a variety of cancers, and its related molecular mechanism affects tumor cell proliferation, invasion, metastasis, and apoptosis including hepatocellular carcinoma, prostate cancer, breast cancer, gastric cancer, gliomas, lung adenocarcinoma, colorectal cancer, ovarian cancer, and endometrial cancer. Clinically, the expression of NCAPG is strongly correlated with N-classification, M-classification, and clinical stage, and NCAPG is valuable for the prognosis of patients with lung adenocarcinoma. In addition, NCAPG can also reduce the sensitivity of tumor cells such as breast cancer to reduce the reaction of the original chemotherapy, so that tumor cells are drug-resistance. In summary, NCAPG can serve as a new diagnosis and treatment target for a variety of cancers, and is also a very promising prognostic marker. Therefore, this review summarizes the critical role of NCAPG in the diagnosis, treatment, and prognosis for various cancers, and the mechanism by which NCAPG plays its pivotal roles.


Assuntos
Adenocarcinoma de Pulmão , Carcinoma Hepatocelular , Neoplasias Hepáticas , Masculino , Humanos , Proteínas de Ciclo Celular/metabolismo , Carcinoma Hepatocelular/patologia , Meiose , Neoplasias Hepáticas/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA