Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Genomics ; 115(4): 110646, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37217085

RESUMO

OBJECTIVE: This study aims to dissect impacts of exosomes-delivered PD-L1 and CTLA-4 siRNAs on colorectal cancer (CRC) progression and immune responses. METHODS: Exosomes containing PD-L1 siRNA and CTLA-4 siRNA were prepared and utilized to treat CRC cells to evaluate their effects. A tumor-bearing mouse model was established for verification. RESULTS: Exosomes containing PD-L1 siRNA and CTLA-4 siRNA repressed malignant features of CRC cells and restrained tumor growth and activated tumor immune responses in vivo. Co-culture of CRC cells treated with exosomes containing PD-L1 siRNA and CTLA-4 siRNA with human CD8+ T cells increased the percentage of CD8+ T cells, decreased the apoptotic rate of CD8+ T cells, elevated IL-2, IFN-γ, and TNF-α expression in cell supernatants, reduced adherent density of CRC cells, augmented the positive rate of CRC cells, and subdued tumor immune escape. CONCLUSION: Exosomes containing PD-L1 siRNA and CTLA-4 siRNA suppressed CRC progression and enhanced tumor immune responses.


Assuntos
Neoplasias Colorretais , Exossomos , Humanos , Animais , Camundongos , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , RNA Interferente Pequeno/genética , Evasão Tumoral , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Antígeno CTLA-4/genética , Antígeno CTLA-4/metabolismo , Exossomos/genética , Exossomos/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , RNA de Cadeia Dupla
2.
J Transl Med ; 21(1): 705, 2023 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-37814346

RESUMO

BACKGROUND AND AIMS: The increasing prevalence of metabolic and cardiovascular diseases poses a significant challenge to global healthcare systems. Regular physical activity (PA) is recognized for its positive impact on cardiovascular risk factors. This study aimed to investigate the relationship between moderate-to-vigorous physical activity (MVPA), sedentary behavior (SB), and abdominal aortic calcification (AAC) using data from the National Health and Nutrition Examination Survey (NHANES). METHODS: The study used data from NHANES participants aged 40 and above during the 2013-2014 cycle. AAC scores were assessed using the Kauppila scoring system, and MVPA and SB were self-reported. Sociodemographic variables were considered, and multivariable linear regression models were used to analyze associations between MVPA, SB, and AAC scores. Subgroup analyses were conducted based on age, sex, BMI, hypertension, and diabetes. RESULTS: The study included 2843 participants. AAC prevalence was higher in older age groups, smokers, and those with diabetes or hypertension. Lower socioeconomic status was associated with higher AAC prevalence. Individuals engaged in any level of MVPA exhibited lower AAC rates compared to inactive individuals. Not engaging in occupational MVPA (ß = 0.46, 95% confidence interval = 0.24‒0.67, p < .001) and prolonged SB (ß = 0.28, 95% confidence interval = 0.04‒0.52, p = .023) were associated with higher AAC scores. However, no significant associations were found for transportation and leisure time MVPA. Subgroup analysis revealed age and hypertension as effect modifiers in the MVPA-AAC relationship. CONCLUSIONS: This study highlights the potential benefits of engaging in occupational MVPA and reducing SB in mitigating AAC scores, particularly among older individuals and those with hypertension.


Assuntos
Diabetes Mellitus , Hipertensão , Humanos , Idoso , Exercício Físico , Inquéritos Nutricionais , Comportamento Sedentário
3.
Genomics ; 114(3): 110319, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35227836

RESUMO

Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) have been proposed as a promising strategy for myocardial infarction (MI). This study aims to explore the mechanism of human umbilical cord MSCs (hucMSCs)-derived EVs loaded with miR-223 on MI. Inflammation, cell biological functions, and fibrosis in vitro were measured. Furthermore, MI rat models were established to verify the role of EVs-miR-223 in vivo. The binding relationship between miR-223 and P53 was confirmed. ChIP assay was utilized to observe the combination of P53 and S100A9. The suppressed fibrosis of cardiomyocytes occurred with cells overexpressing miR-223. MiR-223 contributed to the angiogenesis of HUVECs. P53 was a target gene of miR-223. In vivo, miR-223 relieved myocardial fibrosis and inflammation infiltration, and promoted the angiogenesis in MI rats. HucMSC-derived EVs loaded with miR-223 mitigates MI and promotes myocardial repair through the P53/S100A9 axis, manifesting the underlying therapy values of hucMSC-derived EVs loaded with miR-223 in MI.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , MicroRNAs , Infarto do Miocárdio , Humanos , Ratos , Animais , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Vesículas Extracelulares/metabolismo , Infarto do Miocárdio/genética , Infarto do Miocárdio/terapia , Infarto do Miocárdio/metabolismo , Inflamação/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , Cordão Umbilical/metabolismo , Células-Tronco Mesenquimais/metabolismo , Fibrose
4.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 48(2): 213-220, 2023 Feb 28.
Artigo em Inglês, Zh | MEDLINE | ID: mdl-36999468

RESUMO

OBJECTIVES: Abdominal aortic aneurysm is a pathological condition in which the abdominal aorta is dilated beyond 3.0 cm. The surgical options include open surgical repair (OSR) and endovascular aneurysm repair (EVAR). Prediction of acute kidney injury (AKI) after OSR is helpful for decision-making during the postoperative phase. To find a more efficient method for making a prediction, this study aims to perform tests on the efficacy of different machine learning models. METHODS: Perioperative data of 80 OSR patients were retrospectively collected from January 2009 to December 2021 at Xiangya Hospital, Central South University. The vascular surgeon performed the surgical operation. Four commonly used machine learning classification models (logistic regression, linear kernel support vector machine, Gaussian kernel support vector machine, and random forest) were chosen to predict AKI. The efficacy of the models was validated by five-fold cross-validation. RESULTS: AKI was identified in 33 patients. Five-fold cross-validation showed that among the 4 classification models, random forest was the most precise model for predicting AKI, with an area under the curve of 0.90±0.12. CONCLUSIONS: Machine learning models can precisely predict AKI during early stages after surgery, which allows vascular surgeons to address complications earlier and may help improve the clinical outcomes of OSR.


Assuntos
Injúria Renal Aguda , Aneurisma da Aorta Abdominal , Implante de Prótese Vascular , Procedimentos Endovasculares , Humanos , Aneurisma da Aorta Abdominal/cirurgia , Aneurisma da Aorta Abdominal/complicações , Procedimentos Endovasculares/efeitos adversos , Procedimentos Endovasculares/métodos , Estudos Retrospectivos , Implante de Prótese Vascular/efeitos adversos , Injúria Renal Aguda/epidemiologia , Injúria Renal Aguda/etiologia , Aprendizado de Máquina , Resultado do Tratamento , Complicações Pós-Operatórias/epidemiologia , Complicações Pós-Operatórias/etiologia , Fatores de Risco
5.
Cancer Immunol Immunother ; 71(5): 1075-1091, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-34562123

RESUMO

OBJECTIVE: To investigate the effects of lncRNA MIR155HG and Annexin A2 (ANXA2) on colorectal cancer (CRC) and the mechanism of the MIR155HG/ANXA2 axis. METHODS: The expressions of MIR155HG and ANXA2 in human CRC tissues were analyzed for association with pathological characteristics and prognosis of CRC patients. CRC cell lines (Caco2 and HT29) were used to study the effects of MIR155HG or ANXA2 knockdown on tumor cell behaviors and macrophage polarization as well as the effect of M2 polarization on oxaliplatin resistance of CRC cells. RNA immunoprecipitation, RNA pull-down and dual-luciferase reporter assays were applied to verify the targeting relationships among MIR155HG, miR-650 and ANXA2. Heterotopic xenograft models were established to verify the results of cell experiments. RESULTS: MIR155HG and ANXA2 were highly expressed in CRC tissues/cells and of prognostic values for CRC patients. Knockdown of MIR155HG or ANXA2 suppressed M2 macrophage polarization, and proliferation, migration, invasion and oxaliplatin resistance of CRC cells. MIR155HG competed with ANXA2 for binding miR-650 and can also directly target ANXA2. Knockdown of MIR155HG or ANXA2 also inhibited M2 macrophage polarization and CRC progression in nude mice. CONCLUSION: This study highlighted that MIR155HG, by regulating the miR-650/ANXA2 axis, promotes CRC progression and enhances oxaliplatin resistance in CRC cells through M2 macrophage polarization.


Assuntos
Anexina A2 , Neoplasias Colorretais , MicroRNAs , RNA Longo não Codificante , Animais , Anexina A2/genética , Anexina A2/metabolismo , Células CACO-2 , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/patologia , Resistência a Medicamentos , Regulação Neoplásica da Expressão Gênica , Humanos , Macrófagos/metabolismo , Camundongos , Camundongos Nus , MicroRNAs/genética , MicroRNAs/metabolismo , Oxaliplatina/farmacologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
6.
J Cell Physiol ; 235(3): 2557-2568, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31498440

RESUMO

Glycochenodeoxycholate (GCDA) is closely associated with carcinogenesis and chemoresistance of hepatocellular carcinoma (HCC). Signal transducer and activator of transcription 3 (STAT3), a transcription factor, is involved in various human tumors. Whether GCDA induces chemoresistance through STAT3 and the mechanism of action remains elusive. In this study, we firstly found that the expression level of STAT3 has a positive correlation with chemoresistance of HCC cells. Moreover, GCDA can upregulate the expression of STAT3 protein. Hence, we suspect that GCDA may induce chemoresistance of HCC cells via STAT3. Mechanistically, GCDA stimulates phosphorylation of STAT3 at Ser727 site and mediates pSer727-STAT3 protein to translocate and aggregate in the nucleus, which is important for cell survival. When Ser727 of STAT3 mutated to Asp, the capacity of STAT3 to accumulate in the nucleus was attenuated, STAT3-induced cell survival was impaired and GCDA-induced chemoresistance was abolished. In addition, while activation of extracellular signal-regulated kinase 1/2 (ERK1/2) was inhibited by PD98059, phosphorylation of STAT3 at Ser727 induced by GCDA was suppressed. Taken together, these data demonstrate that GCDA-enhanced survival of liver cancer cells may occur through the activation of STAT3 by phosphorylation at Ser727 site via mitogen-activated protein kinase/ERK1/2 pathway, which may contribute to the progression of human liver cancer and chemoresistance.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Ácido Glicoquenodesoxicólico/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Fator de Transcrição STAT3/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Flavonoides/farmacologia , Fluoruracila/efeitos adversos , Fluoruracila/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Sistema de Sinalização das MAP Quinases/genética , Fosforilação/efeitos dos fármacos
7.
FASEB J ; 33(10): 11396-11410, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31311317

RESUMO

Recent recognition that TGF-ß signaling disruption is involved in the development of aortic aneurysms has led to renewed investigations into the role of TGF-ß biology in the aortic wall. We previously found that the type I receptor of TGF-ß (TGFBR2) receptor contributes to formation of ascending aortic aneurysms and dissections (AADs) induced by smooth muscle cell (SMC)-specific, postnatal deletion of Tgfbr1 (Tgfbr1iko). Here, we aimed to decipher the mechanistic signaling pathway underlying the pathogenic effects of TGFBR2 in this context. Gene expression profiling demonstrated that Tgfbr1iko triggers an acute inflammatory response in developing AADs, and Tgfbr1iko SMCs express an inflammatory phenotype in culture. Comparative proteomics profiling and mass spectrometry revealed that Tgfbr1iko SMCs respond to TGF-ß1 stimulation via robust up-regulation of cyclophilin A (CypA). This up-regulation is abrogated by inhibition of TGFBR2 kinase activity, small interfering RNA silencing of Tgfbr2 expression, or inhibition of SMAD3 activation. In mice, Tgfbr1iko rapidly promotes CypA production in SMCs of developing AADs, whereas treatment with a CypA inhibitor attenuates aortic dilation by 56% (P = 0.003) and ameliorates aneurysmal degeneration (P = 0.016). These protective effects are associated with reduced aneurysm-promoting inflammation. Collectively, these results suggest a novel mechanism, wherein loss of type I receptor of TGF-ß triggers promiscuous, proinflammatory TGFBR2 signaling in SMCs, thereby promoting AAD formation.-Zhou, G., Liao, M., Wang, F., Qi, X., Yang, P., Berceli, S. A., Sharma, A. K., Upchurch, G. R., Jr., Jiang, Z. Cyclophilin A contributes to aortopathy induced by postnatal loss of smooth muscle TGFBR1.


Assuntos
Aorta/metabolismo , Doenças da Aorta/metabolismo , Ciclofilina A/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Animais , Células Cultivadas , Feminino , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta1/metabolismo , Regulação para Cima/fisiologia
8.
Exp Physiol ; 105(1): 160-173, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31553078

RESUMO

NEW FINDINGS: What is the central question of this study? The aim was to identify abdominal aortic aneurysm (AAA)-associated microRNAs and their target genes in AAA using microarray analysis. What is the main finding and its importance? The main finding was that miR-145 and miR-30c-2* were found to be downregulated microRNAs in AAA, which could exert suppressive effects on AAA progression, and that miR-145 might target RAC2, whereas miR-30c-2* might target PIK3CD, IL1B and RAC2. The findings obtained from the study provide an enhanced understanding of microRNA as a therapeutic target to limit AAA. ABSTRACT: The aim of the study was to identify abdominal aortic aneurysm (AAA)-associated microRNAs (miRNAs) and genes potentially contributing to AAA. Differential analysis was performed to screen out differentially expressed genes (DEGs) and miRNAs in expression datasets of AAA-related miRNAs [GSE51226 (mouse)] and genes [GSE51227 (mouse) and GSE7084 (human)]. Then, gene ontology (GO) enrichment analysis of DEGs was compared with aneurysm-related GO to screen out DEGs related to the disease. The target genes of differential miRNAs were predicted and used to construct a miRNA-DEG regulatory network, followed by Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of target genes. Moreover, the protein-protein interaction network of target genes of miRNAs in the core position (hub-miRNA) with AAA-related genes was constructed to screen out hub genes. Finally, the target relationship between hub-miRNAs and their target genes was verified. There were 20 upregulated miRNAs and 20 downregulated miRNAs in AAA screened from the GSE51226 dataset (mouse). In addition, there were 1154 upregulated genes and 821 downregulated genes in the GSE51227 dataset (mouse), of which 246 DEGs were enriched in aneurysm-related GO entries in AAA. miR-145 and miR-30c-2* were the key miRNAs of AAA, both of which were downregulated in AAA and influenced pathways so as to affect AAA by regulating their respective target genes. The disease-related gene ACTA2 was downregulated, whereas DEGs including PIK3CD, IL1B, RAC2 and SELL were upregulated in AAA. Finally, it was proved that miR-145 targeted RAC2 and SELL, whereas miR-30c-2* targeted PIK3CD, IL1B and RAC2. Taken together, miR-145 and miR-30c-2*, downregulated in AAA, could potentially affect AAA, and miR-145 might target RAC2, whereas miR-30c-2* might target PIK3CD, IL1B and RAC2.


Assuntos
Aneurisma da Aorta Abdominal/genética , MicroRNAs/genética , Transcriptoma , Animais , Regulação para Baixo , Ontologia Genética , Redes Reguladoras de Genes , Humanos , Camundongos , Análise em Microsséries , Mapas de Interação de Proteínas , Regulação para Cima
9.
Eur J Vasc Endovasc Surg ; 60(2): 254-263, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32423743

RESUMO

OBJECTIVE: The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signalling pathway plays a pivotal role in abdominal aortic aneurysm (AAA). However, systemic inhibition of this pathway causes serious side effects, thus limiting the clinical use of pan-PI3K inhibitors. In this study, it was hypothesised that the γ subunit of PI3K plays an important role in the PI3K/AKT signalling pathway during AAA, and that specifically targeting PI3Kγ may prevent this process. METHODS: Aortic specimens were collected from AAA patients and organ donors. Furthermore, a classical AAA model in male C57BL/6 mice was created via an intra-aortic porcine pancreatic elastase (PPE) infusion and aortas were collected. A specific PI3Kγ inhibitor, IPI-549, was administered to mice orally. The protein expression level of PI3Kγ was examined by immunohistochemistry and western blotting. The aortic leukocytes were examined by immunohistochemistry and flow cytometry. RESULTS: PI3Kγ protein levels were elevated in the aortas of AAA patients and PPE infused mice. Three color immunofluorescence staining revealed the predominant area of PI3Kγ by T cells and macrophages in aneurysmal aortas. IPI-549 treatment significantly prevented AAA formation in mice. Aortic macrophages, T cells and neo-angiogenesis were significantly reduced in mice treated with IPI-549 compared with vehicle treated PPE infused mice. Flow cytometry analysis also revealed that CD45+ leukocytes and CD45+ F4/80+ macrophages in IPI-549 treated mouse aortas decreased dramatically. Additionally, IPI-549 treatment inhibited the phosphorylation of AKT in experimental aneurysmal lesions. CONCLUSION: Specific inhibition of PI3Kγ limits AAA formation. Targeting PI3Kγ prevents inflammatory cell infiltration through inhibition of AKT phosphorylation in AAA.


Assuntos
Aorta Abdominal/efeitos dos fármacos , Aneurisma da Aorta Abdominal/prevenção & controle , Classe Ib de Fosfatidilinositol 3-Quinase/metabolismo , Isoquinolinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Idoso , Animais , Aorta Abdominal/enzimologia , Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/enzimologia , Aneurisma da Aorta Abdominal/patologia , Estudos de Casos e Controles , Modelos Animais de Doenças , Feminino , Humanos , Isoquinolinas/uso terapêutico , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirazóis/uso terapêutico , Pirimidinas/uso terapêutico , Linfócitos T/efeitos dos fármacos , Linfócitos T/enzimologia
10.
J Cell Physiol ; 234(9): 16592-16600, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30805927

RESUMO

Hepatocellular carcinoma (HCC) is one of the most common malignant tumors in adults. Basic leucine zipper and W2 domains 2 (BZW2) is a member of the basic-region leucine zipper (bZIP) superfamily of transcription factors. Here, we found that BZW2 expression was substantially increased in both human HCC tissues and cell lines, which was correlated with the clinical progression of patients with HCC. Silence of BZW2 in HCC cells by infecting with the lentivirus for delivering BZW2 shRNA (short hairpin RNA), prohibits cell progression, as determined by the suppressed cell proliferation, clonality, invasion, and increased cell apoptosis. Furthermore, overexpression of BZW2 promotes drug resistance of HCC cells, as shown by the attenuated suppression of cell viability and invasion following rapamycin (RAPA) treatment. Mechanistically, overexpression (or silence) of BZW2 in HCC cells significantly stimulates (or decreases) the activation of the PI3K/AKT/mTOR signaling pathway, which is responsible for HCC progression. Thus, increased BZW2 expression in HCC can induce HCC progression and drug resistance via stimulating the PI3K/AKT/mTOR pathway, which may represent a new therapeutic target for HCC.

11.
J Cell Physiol ; 234(9): 15934-15940, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30741422

RESUMO

Nicotine is a major carcinogen in cigarettes, which can enhance cell proliferation and metastasis and increase the chemoresistance of cancer cells. Our previous data found that nicotine promotes cell survival in lung cancer by affecting the expression of antiapoptotic protein Mcl-1, suggesting that the Mcl-1 may be a therapeutic target for patients with lung cancer. In this study, we found that the effects of drug resistance on nicotine-induced lung cancer cell lines were shown to influence the phosphorylation of Mcl-1. Moreover, nicotine induces Mcl-1 phosphorylation exclusively at the T163 site, which results in enhancement of the antiapoptotic activity of Mcl-1 and increased cell survival. Meanwhile, nicotine can reduce the sensitivity of H1299 cells to CDDP via enhancement of the binding of Mcl-1 to Bak, which inhibits the proapoptotic effect of Bak and ultimately leads to increased survival and drug resistance of lung cancer cells. Thus, nicotine-induced cell survival and chemoresistance may occur in a mechanism by stimulating Mcl-1 phosphorylation and its interaction with Bak, which may contribute to improving the efficacy of chemotherapy in the treatment of human lung cancer.

12.
J Cell Physiol ; 234(7): 10899-10906, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30548625

RESUMO

OBJECTIVE: Glycochenodeoxycholate acid (GCDA) is a toxic component in bile salts. It plays an important role in the development and progression of liver cancer. In this study, we investigated the underlying mechanism of GCDA in hepatocarcinogenesis and chemotherapy resistance. MATERIALS AND METHODS: Cell proliferation was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and clonality by Ki-67 and colony-formation assay. Apoptosis was examined by flow cytometry. Real-time polymerase chain reaction (PCR) and western blot analysis were used to measure messenger RNA and protein levels, respectively. Short hairpin RNA was used to silence signal transducer and activator of transcription 3 (Stat3) expression. RESULTS: Bile salts (GCDA) promoted the proliferation of hepatocellular carcinoma (HCC) cells (HepG2 and QGY-7703), and GCDA treatment reduced the chemosensitivity of 5-fluorouracil (5FU) in HepG2 and QGY-7703 cells. GCDA upregulated the expression of antiapoptosis proteins Mcl-1/Survivin/Bcl-2. GCDA had no discernible effect on basal protein level or subcellular localization of phosphorylated Stat3. 5FU increased the apoptosis of HepG2 cells with silenced Stat3 expression, but GCDA-induced chemoresistance was not reversed. CONCLUSIONS: GCDA-reduced HCC cell chemosensitivity may occur by upregulating antiapoptosis proteins Mcl-1/Survivin/Bcl-2. Stat3 may be a target for enhancing the chemosensitivity of hepatocellular carcinoma cells, but GCDA-induced chemoresistance is independent of Stat3.


Assuntos
Carcinoma Hepatocelular/patologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ácido Glicoquenodesoxicólico/toxicidade , Neoplasias Hepáticas/patologia , Antimetabólitos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fluoruracila/farmacologia , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Survivina/genética , Survivina/metabolismo
13.
Nanomedicine ; 15(1): 86-97, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30244085

RESUMO

Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related deaths worldwide. Current therapies present significant limitations. Triptolide (TP) is highly effective against multiple cancers including HCC. However, high toxicity, low water solubility, and unknown therapeutic targets limit its clinical application. Herein, we designed galactosylated-chitosan-TP-nanoparticles (GC-TP-NPs) with high drug loading capacities for targeted delivery to HCC. In addition to a sustained release pattern, an efficient asialoglycoprotein receptor mediated cellular uptake in vitro, and high liver tumor accumulation in vivo, GC-TP-NPs showed lower systemic and male reproductive toxicities than free TP. Importantly, GC-TP-NPs retained the anti-cancer activities of the free TP, exerting the same pro-apoptotic and anti-proliferative effects on HCC cells in vitro, and displayed higher efficacies in reducing tumor sizes in vivo. Further investigation revealed that GC-TP-NPs induced cancer cell apoptosis via blocking TNF/NF-κB/BCL2 signaling. Collectively, GC-TP-NP represents a promising candidate in halting liver cancer progression while minimizing systemic toxicity.


Assuntos
Carcinoma Hepatocelular/prevenção & controle , Quitosana/química , Diterpenos/administração & dosagem , Galactose/química , Neoplasias Hepáticas/prevenção & controle , Nanopartículas/administração & dosagem , Fenantrenos/administração & dosagem , Reprodução , Animais , Antineoplásicos Alquilantes/administração & dosagem , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/farmacocinética , Apoptose , Carcinoma Hepatocelular/patologia , Proliferação de Células , Diterpenos/química , Diterpenos/farmacocinética , Compostos de Epóxi/administração & dosagem , Compostos de Epóxi/química , Compostos de Epóxi/farmacocinética , Humanos , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Nus , Nanopartículas/química , Fenantrenos/química , Fenantrenos/farmacocinética , Transdução de Sinais , Distribuição Tecidual , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Genesis ; 55(9)2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28845554

RESUMO

The Myh11-CreERT2 mouse line (Cre+ ) has gained increasing application because of its high lineage specificity relative to other Cre drivers targeting smooth muscle cells (SMCs). This Cre allele, however, was initially inserted into the Y chromosome (X/YCre+ ), which excluded its application in female mice. Our group established a Cre+ colony from male ancestors. Surprisingly, genotype screening identified female carriers that stably transmitted the Cre allele to the following generations. Crossbreeding experiments revealed a pattern of X-linked inheritance for the transgene (k > 1000), indicating that these female carries acquired the Cre allele through a mechanism of Y to X chromosome translocation. Further characterization demonstrated that in hemizygous X/XCre+ mice Cre activity was restricted to a subset arterial SMCs, with Cre expression in arteries decreased by 50% compared to X/YCre+ mice. This mosaicism, however, diminished in homozygous XCre+ /XCre+ mice. In a model of aortic aneurysm induced by a SMC-specific Tgfbr1 deletion, the homozygous XCre+ /XCre+ Cre driver unmasked the aortic phenotype that is otherwise subclinical when driven by the hemizygous X/XCre+ Cre line. In conclusion, the Cre allele carried by this female mouse line is located on the X chromosome and subjected to X-inactivation. The homozygous XCre+ /XCre+ mice produce uniform Cre activity in arterial SMCs.


Assuntos
Cadeias Pesadas de Miosina/genética , Translocação Genética , Cromossomo X/genética , Cromossomo Y/genética , Alelos , Animais , Feminino , Hemizigoto , Homozigoto , Integrases/genética , Integrases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mosaicismo , Músculo Liso Vascular/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Fenótipo , Transgenes , Doenças Vasculares/genética , Doenças Vasculares/patologia
15.
Tumour Biol ; 39(7): 1010428317712102, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28681695

RESUMO

In cholestasis, increases in bile acid levels result in the generation of reactive oxygen species and the induction of DNA damage and mutation. It is believed that bile acid accumulation is associated with liver tumorigenesis. However, the mechanism that underpins this phenomenon remains to be elucidated. Mcl-1, which is overexpressed in hepatic cells, is a pro-survival member of the Bcl-2 family. In this study, we observed that Mcl-1 potently suppresses the repair of bile acid-induced abasic (apurinic/apyrimidinic) sites in DNA lesions. Upon exposure of hepatic cells to glycochenodeoxycholate, one of the major conjugated human bile acids, we observed an increase in AP site accumulation along with induction of poly(ADP-ribose) polymerase and XRCC1 ( X-Ray Repair Cross Complementing 1). In addition, accumulation of Mcl-1 was observed in the nuclei of QGY-7703 cells in response to glycochenodeoxycholate stimulation. Knockdown of endogenous Mcl-1 by RNA interference significantly accelerated the repair of DNA lesions in glycochenodeoxycholate-treated cells. However, unlike XRCC1, poly(ADP-ribose) polymerase was induced following Mcl-1 knockdown. Conversely, poly(ADP-ribose) polymerase suppression was observed following glycochenodeoxycholate treatment of cells overexpressing Mcl-1. Moreover, AP-site counting analyses revealed that DNA repair activity was enhanced in cells overexpressing poly(ADP-ribose) polymerase under glycochenodeoxycholate stress conditions. It is well known that poly(ADP-ribose) polymerase plays a crucial role in the base excision repair pathway. Thus, our findings suggest that Mcl-1 suppresses base excision repair by inhibiting poly(ADP-ribose) polymerase induction following glycochenodeoxycholate-induced DNA damage. These results potentially explain how bile acid accumulation results in genetic instability and carcinogenesis.


Assuntos
Colestase/genética , Proteínas de Ligação a DNA/genética , Neoplasias Hepáticas/genética , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Poli(ADP-Ribose) Polimerases/genética , Ácido Apurínico/genética , Ácidos e Sais Biliares/normas , Ácidos e Sais Biliares/toxicidade , Colestase/metabolismo , Colestase/patologia , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Ácido Glicoquenodesoxicólico/toxicidade , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Espécies Reativas de Oxigênio/metabolismo , Proteína 1 Complementadora Cruzada de Reparo de Raio-X
16.
Tumour Biol ; 37(7): 9875-86, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26812694

RESUMO

The objective of this study was to investigate the expression, proliferation, and apoptosis function of long-chain non-coding RNA maternally expressed gene 3 (MEG3) and antisense non-coding RNA at the INK4 locus (ANRIL) in gallbladder cancer (GBC) tissues. GBC tissues and adjacent normal samples were collected from 84 patients from January 2008 to June 2010. Empty vector, pcDNA-MEG3, and pcDNA-ANRIL vectors were transfected into GBC-SD and QBC939 cells. An MTT assay, real-time quantitative polymerase chain reaction (RT-qPCR), flow cytometry, Western blotting, and immunohistochemistry were applied. The effects of MEG3 and ANRIL were also verified in mice. Compared with normal tissues, the expression of MEG3 was significantly lower in GBC tissues, whereas the expression of ANRIL was significantly higher (both P < 0.05). The overexpression of MEG3 and underexpression of ANRIL were significantly associated with GBC prognosis (both P < 0.05). The expressions of MEG3 and ANRIL were higher in pcDNA-MEG3 and pcDNA-ANRIL-transfected cells than in empty vector-transfected cells in vitro (both P < 0.05). Most of the pcDNA-MEG3-transfected cells were in the G0-G1 phase, which showed reduced cell activity and clone counts and increased p53 and decreased cyclin D1, whereas the pcDNA-ANRIL-transfected cells were mostly in the S phase and showed contrasting behavior. Mice injected with pcDNA-MEG3-transfected cells had smaller and lighter tumors, decreased ki-67 levels, and increased caspase 3 levels, whereas those injected with pcDNA-ANRIL showed contrasting results (all P < 0.05). MEG3 can inhibit the proliferation of GBC cells and promote apoptosis, whereas ANRIL can improve the proliferation of gallbladder cells and inhibit apoptosis. Collectively, our results suggest that therapeutic strategies directed toward upregulating MEG3 and downregulating ANRIL may be clinically relevant for the inhibition of GBC deterioration.


Assuntos
Adenocarcinoma/genética , Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/genética , Neoplasias da Vesícula Biliar/genética , RNA Longo não Codificante/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Apoptose , Western Blotting , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Estudos de Casos e Controles , Proliferação de Células , Feminino , Seguimentos , Neoplasias da Vesícula Biliar/metabolismo , Neoplasias da Vesícula Biliar/patologia , Humanos , Técnicas Imunoenzimáticas , Metástase Linfática , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Invasividade Neoplásica , Estadiamento de Neoplasias , Prognóstico , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
17.
PLoS One ; 19(2): e0296729, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38335213

RESUMO

BACKGROUND: Rupture of abdominal aortic aneurysm (rAAA) is a fatal event in the elderly. Elevated blood pressure and weakening of vessel wall strength are major risk factors for this devastating event. This present study examined whether the expression profile of mechanosensitive genes correlates with the phenotype and outcome, thus, serving as a biomarker for AAA development. METHODS: In this study, we identified mechanosensitive genes involved in AAA development using general bioinformatics methods and machine learning with six human datasets publicly available from the GEO database. Differentially expressed mechanosensitive genes (DEMGs) in AAAs were identified by differential expression analysis. Molecular biological functions of genes were explored using functional clustering, Protein-protein interaction (PPI), and weighted gene co-expression network analysis (WGCNA). According to the datasets (GSE98278, GSE205071 and GSE165470), the changes of diameter and aortic wall strength of AAA induced by DEMGs were verified by consensus clustering analysis, machine learning models, and statistical analysis. In addition, a model for identifying AAA subtypes was built using machine learning methods. RESULTS: 38 DEMGs clustered in pathways regulating 'Smooth muscle cell biology' and 'Cell or Tissue connectivity'. By analyzing the GSE205071 and GSE165470 datasets, DEMGs were found to respond to differences in aneurysm diameter and vessel wall strength. Thus, in the merged datasets, we formally created subgroups of AAAs and found differences in immune characteristics between the subgroups. Finally, a model that accurately predicts the AAA subtype that is more likely to rupture was successfully developed. CONCLUSION: We identified 38 DEMGs that may be involved in AAA. This gene cluster is involved in regulating the maximum vessel diameter, degree of immunoinflammatory infiltration, and strength of the local vessel wall in AAA. The prognostic model we developed can accurately identify the AAA subtypes that tend to rupture.


Assuntos
Aneurisma da Aorta Abdominal , Ruptura Aórtica , Humanos , Idoso , Aneurisma da Aorta Abdominal/metabolismo , Fatores de Risco , Aorta/metabolismo , Prognóstico , Biomarcadores , Ruptura Aórtica/genética
18.
Front Cardiovasc Med ; 10: 1044560, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36844737

RESUMO

Purpose: To review the association between central obesity and abdominal aortic aneurysm (AAA). Materials and methods: The PubMed, Web of Sciences, Embase, The China national knowledge infrastructure (CNKI), and Cochrane Library were searched up to April 30, 2022. Researches includes investigation of the relationship between central obesity markers and AAA. Included studies must use recognized measures of central obesity, i.e., waist circumference (WC) and waist-to-hip ratio (WHR), or use imaging techniques to calculate abdominal fat distribution, such as computed tomography (CT) imaging. Results: Eleven clinical researches were identified of which eight discussed the association between physical examination and AAA, and three studies mainly focused on abdominal fat volume (AFV). Seven researches concluded that there was a positive correlation between markers of central obesity and AAA. Three studies found no significant link between markers of central obesity and AAA. One of the remaining studies reported different results for each sex. Three studies pooled in a meta-analysis identified correlation between central obesity and AAA presence (RR = 1.29; 95% confidence interval, 1.14-1.46). Conclusion: Central obesity plays a role in the risk of AAA. Standardized central obesity markers may be predictors of AAA. However, there was no association between abdominal fat volume and AAA. Additional relevant evidence and specific mechanisms warrant further study. Systematic review registration: https://www.crd.york.ac.uk/prospero/display_record.php?IDCRD42022332519, identifier CRD42022332519.

19.
Transl Oncol ; 32: 101655, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36963204

RESUMO

BACKGROUND: Glypican-1 (GPC1) is overexpressed in several tumors, and GPC1+ exosomes have shown the potential to predict early colorectal cancer (CRC). However, the mechanisms underlying the enrichment and action of GPC1+ exosomes in CRC remain unknown. METHODS: The expression of slit guidance ligand 2 (SLIT2), hypoxia-inducible factor (HIF)-1α/2α, and GPC1 in clinical CRC tissues was detected using immunohistochemistry and western blot. Exosomes were isolated from the supernatants of CRC cell cultures. The effects of SLIT2, hypoxia, heparin, and phospholipase C (PLC) on exosomal GPC1 expression and GPC1+ exosome enrichment in CRC cells were analyzed with western blot and flow cytometry. CRC cell proliferation was assessed with MTT and colony formation assays. Co-immunoprecipitation was used to detect the binding of GPC1 and SLIT2 in SW480 cells. Nude mice were subcutaneously inoculated with SW480 cells with different treatments. The Wnt signaling was detected. RESULTS: SLIT2 was poorly expressed and GPC1, HIF-1α, and HIF-2α were highly expressed in human CRC tissues. SLIT2 in CRC cells inhibited GPC1+ exosome enrichment and exosomal GPC1 expression. PLC and heparin increased GPC1+ exosome enrichment in CRC cells in a concentration-dependent manner. Hypoxia increased the enrichment of GPC1+ exosomes in CRC cells depending on HIF-2α expression. GPC1+ exosomes stimulated CRC cell proliferation and xenograft tumor growth through activation of Wnt signaling. CONCLUSIONS: GPC1+ exosome enrichment is related to PLC and heparin. Hypoxia increases the enrichment of GPC1+ exosomes in CRC cells by activating HIF-2α and downregulating SLIT2. GPC1+ exosomes further drive CRC progression by activating Wnt signaling.

20.
Nutrients ; 14(16)2022 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-36014855

RESUMO

Accumulating evidence in recent years has demonstrated the important role of gut microbiota in maintaining cardiovascular function. However, their functions in abdominal aortic aneurysm (AAA) are largely unexplored. In this study, we established a porcine pancreatic elastase-infused experimental AAA mouse model and explored gut microbiota modulation using 16S rDNA sequencing. Here, we found that a significant alteration to gut microbiota composition and function occurred in AAA. The functional change in the gut microbiome revealed dysregulated biosynthesis metabolism and transport of spermidine in AAA. Furthermore, exogenous spermidine was administrated via drinking water and attenuated the progression of experimental AAA disease, which supports our recent study that spermidine alleviates systemic inflammation and AAA. These effects were associated with remitted gut microbiota dysbiosis and metabolism in AAA progression as demonstrated by 16S rDNA gene analysis. In addition, several bacterial florae, such as Bacteroides, Parabacteroides and Prevotella, were identified to be associated with the progression of AAA. Our results uncovered altered gut microbial profiles in AAA and highlighted the potential therapeutic use of spermidine in the treatment of gut microbiota dysbiosis and AAA.


Assuntos
Aneurisma da Aorta Abdominal , Microbioma Gastrointestinal , Animais , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/tratamento farmacológico , DNA Ribossômico , Modelos Animais de Doenças , Disbiose/tratamento farmacológico , Disbiose/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Espermidina/farmacologia , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA