Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Cell Mol Med ; 28(2): e18048, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37986543

RESUMO

Intervertebral disc degeneration (IVDD) is a common chronic musculoskeletal disease that causes chronic low back pain and imposes an immense financial strain on patients. The pathological mechanisms underlying IVDD have not been fully elucidated. The development of IVDD is closely associated with abnormal epigenetic changes, suggesting that IVDD progression may be controlled by epigenetic mechanisms. Consequently, this study aimed to investigate the role of epigenetic regulation, including DNA methyltransferase 3a (DNMT3a)-mediated methylation and peroxisome proliferator-activated receptor γ (PPARγ) inhibition, in IVDD development. The expression of DNMT3a and PPARγ in early and late IVDD of nucleus pulposus (NP) tissues was detected using immunohistochemistry and western blotting analyses. Cellularly, DNMT3a inhibition significantly inhibited IL-1ß-induced apoptosis and extracellular matrix (ECM) degradation in rat NP cells. Pretreatment with T0070907, a specific inhibitor of PPARγ, significantly reversed the anti-apoptotic and ECM degradation effects of DNMT3a inhibition. Mechanistically, DNMT3a modified PPARγ promoter hypermethylation to activate the nuclear factor-κB (NF-κB) pathway. DNMT3a inhibition alleviated IVDD progression. Conclusively, the results of this study show that DNMT3a activates the NF-κB pathway by modifying PPARγ promoter hypermethylation to promote apoptosis and ECM degradation. Therefore, we believe that the ability of DNMT3a to mediate the PPARγ/NF-κB axis may provide new ideas for the potential pathogenesis of IVDD and may become an attractive target for the treatment of IVDD.


Assuntos
Degeneração do Disco Intervertebral , Disco Intervertebral , Núcleo Pulposo , Animais , Humanos , Ratos , DNA Metiltransferase 3A , Epigênese Genética , Disco Intervertebral/patologia , Degeneração do Disco Intervertebral/patologia , Metilação , NF-kappa B/metabolismo , Núcleo Pulposo/patologia , PPAR gama/genética , PPAR gama/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais
2.
Immunol Invest ; 52(3): 343-363, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36762677

RESUMO

BACKGROUND: Programmed death ligand 1 (PD-L1) is expressed in hepatocellular carcinoma (HCC) cells. PD-L1 function and structure are regulated through glycosylation and various signaling pathways. However, the relationship between Pseudomonas aeruginosa mannose sensitive hemagglutinin (PA-MSHA), glycosylation and PD-L1 warrants further study. In this study, we investigated the effects of PA-MSHA on the regulation of mannosyl and N-glycosylation to identify the mechanisms underlying its function. METHODS: PD-L1, ß-catenin, c-Myc, mannosyl, MGAT1 and mannosidase II in HCC were identified by postoperative specimens from the HCC cohort with immunohistochemistry and immunofluorescence. PA-MSHA was used to suppress tumor progression. Alterations to the expression of PD-L1, ß-catenin, c-Myc, MGAT1, and mannosidase II at the gene and protein levels were detected by qRT-PCR and Western blot analysis. Soluble PD-L1 (sPD-L1) were detected using enzyme-linked immunosorbent assay. RESULTS: Mannosyl and mannosidase II expression levels increased, whereas those of MGAT1 decreased in the HCC cells. The glycosylation-related pathway proteins, namely, ß-catenin, c-Myc and PD-L1, had increased expression levels. Moreover, proliferation in the HCC cells was inhibited after PA-MSHA treatment, PD-L1 function was significantly inhibited. Transmission electron microscopy showed that PA-MSHA penetrated into the HCC cytoplasm through the cytomembrane, resulting in apoptosis. Here, PA-MSHA significantly reduced sPD-L1 expression levels in the tumor cells. CONCLUSIONS: PA-MSHA plays the role of a lectin, affecting receptors on the cytomembrane. This strain inhibits mannosyl by suppressing ß-catenin signaling. We hypothesized that PA-MSHA suppresses PD-L1 by: 1. Inhibiting the glycosylation process; and 2. Suppressing ß-catenin and c-Myc, thereby reducing the transcription of this protein.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Pseudomonas aeruginosa , Humanos , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Idoso , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/terapia , Animais , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Xenoenxertos , Antígeno B7-H1/metabolismo , Linhagem Celular Tumoral , Pseudomonas aeruginosa/classificação , Pseudomonas aeruginosa/fisiologia , Transplante de Neoplasias , Glicosilação , Transdução de Sinais , Imunoterapia , Lectinas/metabolismo
3.
Neoplasma ; 70(1): 1-14, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36129834

RESUMO

The changes in cell homeostasis in the tumor microenvironment may affect the development of colorectal cancer (CRC). Genomic instability is an important factor. Persistent genomic instability leads to epigenetic changes, and mutations are a major factor in the progression of CRC. Based on these mechanisms, it is reasonable to link poly (ADP-ribose) polymerase (PARP) with the treatment of CRC. PARP is mainly involved in DNA repair, which has an essential role in the DNA damage response and prevention of DNA damage, and maintains oxidation and superoxide redox homeostasis in the intracellular environment of the tumor. This article reviews the latest research progress on PARP and PARP inhibitors (PARPi) in CRC. It mainly includes molecular mechanisms, immunity, clinical trials, and combination strategies of CRC. The research of PARPi in CRC has broad prospects, and the combinations with other drugs are the main research direction in the future.


Assuntos
Neoplasias Colorretais , Inibidores de Poli(ADP-Ribose) Polimerases , Humanos , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Dano ao DNA , Poli(ADP-Ribose) Polimerases/genética , Instabilidade Genômica , Combinação de Medicamentos , Neoplasias Colorretais/genética , Microambiente Tumoral
4.
Acta Biochim Biophys Sin (Shanghai) ; 55(4): 649-660, 2023 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-36786074

RESUMO

Tumour cells change their metabolic patterns to support high proliferation rates and cope with oxidative stress. The lncRNA ELFN1-AS1 is highly expressed in a wide range of cancers and is essential to the proliferation and apoptosis of tumour cells. Nevertheless, its function in the metabolic reprogramming of tumour cells is unclear. Here we show that ELFN1-AS1 promotes glucose consumption as well as lactate and NADPH production. Database searching, bioinformatics analysis, RNA immunoprecipitation (RIP) and RNA pull-down assays show that ELFN1-AS1 enhances glucose-6-phosphate dehydrogenase ( G6PD) expression and activates the pentose phosphate pathway (PPP) by promoting TP53 degradation. In addition, luciferase reporter assay and chromatin immunoprecipitation (ChIP) show that YY1 binds to the ELFN1-AS1 promoter to promote transcriptional activation of ELFN1-AS1. Consistent with the in vitro experiments, knockdown of ELFN1-AS1 impedes the growth of tumours transplanted into mice by inhibiting the expression of G6PD. In conclusion, this study reveals that ELFN1-AS1 activates the PPP, and validates the regulatory role of the YY1/ ELFN1-AS1/ TP53/ G6PD axis in colorectal cancer.


Assuntos
Neoplasias Colorretais , MicroRNAs , RNA Longo não Codificante , Animais , Camundongos , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Neoplasias Colorretais/metabolismo , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Humanos
5.
Jpn J Clin Oncol ; 52(4): 331-345, 2022 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-35106596

RESUMO

BACKGROUND: Till now, no experiment has been performed to detect programmed death ligand 1 (PD-L1)/programmed death 1 (PD-1), soluble PD-L1/soluble PD-1 simultaneously in perioperative patients of gastric carcinoma. Our experiment aims at determining the clinical significance and possible mechanism of soluble PD-L1/soluble PD-1 in gastric carcinoma. METHODS: Thirty patients undergone gastrectomy were selected as the experimental group. Tissue's programmed death ligand 1 and peripheral programmed death 1 were detected using immunofluorescence and flow cytometry. Soluble PD-L1 and soluble PD-1 were detected using enzyme-linked immunosorbent assay. RESULTS: First, preoperative programmed death 1 was higher than control group and decreased to normal post-operatively. Preoperatively ,elevated levels of programmed death 1 on cluster of differentiation (CD)4 T cells indicated less lymphatic metastasis (P < 0.01) and small tumor volume (P < 0.01); elevated programmed death 1 of CD8 T cells indicated big tumor volume (P < 0.01) and well histological differentiation (P < 0.01). Second, preoperative soluble PD-L1 and soluble PD-1 are lower than in control group. Post-operatively, the soluble PD-1 rose to normal, but the soluble PD-L1 showed no change. Third, programmed death ligand 1 can be observed in carcinoma tissue. Fourth, the area under the curve of soluble PD-1 (0.675) for diagnosis was worse than that of soluble PD-L1 (0.885). Kaplan-Meier analysis showed that soluble PD-1 < 245.26 pg/ml in post-operative serum predicted a poor prognosis (overall survival percentage: 60%) at 2 years (P < 0.05). Multivariate analysis revealed that carcinoembryonic antigen (>5 ng/l) and soluble PD-1 after gastrectomy (>245.26 pg/ml) were independent prognostic factors for overall survival (hazard ratio: 20.812, 95% confidence interval: 1.217-355.916, P = 0.036; hazard ratio: 0.028, 95% confidence interval: 0.001-0.786, P = 0.036, respectively). CONCLUSIONS: We propose that soluble PD-1 combined with programmed death ligand 1 are effective not only in protecting T cells from the adhesion by programmed death ligand 1 but also in preventing the occurrence and the development of tumor as well. Through multivariate analysis, we found that soluble PD-1 was an independent protective factor for post-operative prognosis of gastric carcinoma patients, which indirectly verified the vital function of soluble PD-1. Soluble PD-1 might be promising predictive biomarkers for the diagnosis and prognosis of gastric carcinoma patients.


Assuntos
Antígeno B7-H1 , Carcinoma , Receptor de Morte Celular Programada 1 , Neoplasias Gástricas , Antígeno B7-H1/metabolismo , Biomarcadores Tumorais/metabolismo , Humanos , Período Perioperatório , Prognóstico , Receptor de Morte Celular Programada 1/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Neoplasias Gástricas/cirurgia
6.
Neoplasma ; 69(3): 491-503, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35103480

RESUMO

With the continuous development of medical science and technology, the medical community's understanding of the disease is constantly updated, just as strategies for treating malignant tumors are constantly updated. New diagnoses, follow-up indicators, and treatment plan formulations need more evidence to be supported. To date, radical surgical resection is still the preferred treatment for advanced digestive system malignancies, and combination therapy including chemotherapy and targeted therapy before or after surgery is aimed at improving the prognosis and quality of life of patients. However, if tumor recurrence, metastasis, chemotherapy, and drug resistance to targeted agents after surgery prevent the achievement of the desired therapeutic effect, and if neoadjuvant chemotherapy and targeted therapy cannot reduce the staging of the tumor, surgery cannot be performed. These are huge problems that we face now and will continue to face for some time. Relevant scientific data and evidence have been produced to explain unsatisfactory efficacy, such as epithelial-mesenchymal transformation, the tumor microenvironment, extracellular matrix proteins, cancer-related fibroblasts, and other factors that may be related to tumor progression and poor therapeutic effects. An extracellular matrix protein, periostin (POSTN), influences the above factors and has received multidisciplinary attention. In this paper, periostin and digestive system-related tumors are reviewed, and the production, mechanism of action, drug resistance correlation analysis, and coping strategies of periostin are summarized to further understand its characteristics. This work provides evidence for potential therapeutic targets for digestive system tumors in the future.


Assuntos
Antineoplásicos , Qualidade de Vida , Antineoplásicos/farmacologia , Transição Epitelial-Mesenquimal , Humanos , Prognóstico , Microambiente Tumoral
7.
Cell Cycle ; 21(11): 1140-1152, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35239447

RESUMO

Cancers continue to have high incidence and mortality rates worldwide. Therefore, cancer control remains the main public health goal. Growing research evidence suggests that phospholysine phosphohistidine inorganic pyrophosphate phosphatase (LHPP) plays an important role in inhibiting tumor cell progression. It has been reported in the literature that LHPP is expressed at low levels in tumor tissues and cells and that patients with low LHPP expression have a poorer prognosis. Functional studies have shown that LHPP can inhibit tumor cell proliferation, metastasis, and apoptosis by affecting different target genes. In addition, researchers have used iDPP nanoparticles to deliver LHPP plasmids to treat tumors, demonstrating the great potential of LHPP plasmids for cancer therapy. In our review, we highlight the biological functions and important downstream target genes of LHPP in tumors, providing a theoretical basis for the treatment of human cancers. Although not thoroughly studied in terms of tumor mechanisms, LHPP still represents a promising and effective anticancer drug target.


Assuntos
Histidina , Neoplasias , Proliferação de Células , Humanos , Pirofosfatase Inorgânica/genética , Pirofosfatase Inorgânica/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/genética , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo
8.
Cell Cycle ; 21(24): 2563-2574, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35912542

RESUMO

Colorectal cancer (CRC) is now the third most prevalent tumor and one of the deadliest cancers worldwide, with an increasing prevalence every year. Therefore, we urgently need to understand the mechanisms regulating the progression of colorectal cancer and find potential diagnostic biomarkers. In this study, we performed an analysis using the TCGA and GEO databases to find a molecular biomarker for the diagnosis of CRC, namely CTPS1. The results of this analysis revealed that CTPS1 could promote tumor proliferation and metastasis. Furthermore, bioinformatics analysis revealed that CTPS1 promoted CRC progression through cell cycle and p53 pathways. Further investigation demonstrated that CTPS1 might be involved in the regulation of CCNB1, RRM2, GTSE1, CDK2 and CHEK2 genes. Moreover, PCR confirmed that CTPS1 regulated GTSE1 and CDK2 molecules. Then, western blot was used to verify that CTPS1 promoted the expression of GTSE1 and CDK2 by inhibiting the expression of p53. In summary, we identified an important diagnostic biomarker for CRC, namely CTPS1, and its importance was validated at the cellular level. These results suggest that CTPS1 could serve as a candidate biomarker for CRC and CTPS1 inhibitors may be a potential treatment for CRC.


Assuntos
Neoplasias Colorretais , Proteína Supressora de Tumor p53 , Humanos , Movimento Celular/genética , Proliferação de Células/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Neoplasias Colorretais/patologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Proteínas Associadas aos Microtúbulos/metabolismo
9.
Front Oncol ; 11: 667904, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34123835

RESUMO

Hepatocellular carcinoma (HCC), one of the most common tumors worldwide, has the fifth highest mortality rate, which is increasing every year. At present, many studies have revealed that immunotherapy has an important effect on many malignant tumors. The main purpose of our research was to verify and establish a new immune-related lncRNA model and to explore the potential immune mechanisms. We analysed the pathways and mechanisms of immune-related lncRNAs by bioinformatics analysis, screened key lncRNAs based on Cox regression analysis, and determined the characteristics of the immune-related lncRNAs. On this basis, a predictive model was established. Through a comparison of specificity and sensitivity, we found that the constructed model was superior to the known markers of HCC. Then, the cell types were identified by the relative subgroup (CIBERSORT) algorithm for RNA transcripts. A signature model was eventually constructed, and we proved that it was a survival factor for HCC. Moreover, five kinds of immune cells were significantly positively correlated with the signature. The results indicated that these five kinds of lncRNAs may be related to the immune infiltration of hepatocellular carcinoma. To verify these findings, we selected the top coexpressed lncRNA, AC099850.3, for further study. We found that AC099850.3 could promote the migration and proliferation of hepatocellular carcinoma cells in vitro. RT-PCR experiments found that AC099850.3 could promote the expression of the cell cycle molecules BUB1, CDK1, PLK1, and TTK, and western blotting to prove that the expression of the molecules CD155 and PD-L1 was inhibited in the interference group. In conclusion, we used five kinds of immune-related lncRNAs to construct prognostic signatures to explore the mechanism, which provides a new way to study therapies for HCC.

10.
Front Pharmacol ; 11: 608218, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33628179

RESUMO

Oxovanadium complexes, particularly vanadyl (IV) derivatives with hybrid ligands of Schiff base and polypyridyl, have been demonstrated to possess great anticancerous therapeutic efficacy. However, most of the studies on the activity of these oxovanadium complexes have mainly focused on in vitro studies, and animal studies in vivo are extremely scarce. Based on the antitumor test results of four novel oxovanadium complexes in our previous work, this work further conducted a comprehensive antitumor activity study in vitro and in vivo on VO(hntdtsc)(NPIP), which owned the strongest inhibitory activity in vitro on multiple tumor cell proliferation. The cellular mechanism study suggested that VO(hntdtsc)(NPIP) inhibited the cell proliferation via arresting the cell cycle at G0/G1 phase through the p16-cyclin D1-CDK4-p-Rb pathway and inducing cell apoptosis through mitochondrial-dependent apoptosis pathway on HeLa cells. Inconsistent with the effects in vitro, VO(hntdtsc)(NPIP) significantly inhibited the growth of tumor and induced the apoptosis of cancer cells in mice xenograft models according to the results of nude mice in vivo image detection, H&E pathological examination, and immunohistochemical detection of p16/Ki-67 protein expression. Collectively, all the results, particularly studies in vivo, demonstrated that VO(hntdtsc)(NPIP) hold a potential to be the lead compound and further to be an anticervical cancer drug.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA