Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Heliyon ; 10(15): e35303, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-39170313

RESUMO

Glioma is one of the prevalent malignancies, and identifying therapeutic targets for glioma is highly important. Findings of current study revealed that inositol-trisphosphate 3-kinase A (ITPKA) was found abnormally over expressed and thereby exhibited poor prognosis with glioma. Extensive academic research has meticulously elucidated ITPKA's pivotal role in enhancing glioma cell proliferation and invasion, highlighting its significance in oncogenic pathways and cellular dynamics specific to aggressive brain tumors. Inhibiting the ITPKA has wide scope to reduce the tumorigenicity in gliomas in vivo. We also noticed that ITPKA interacts with PYCR1 and phosphorylates serine 29 of PYCR1. Phosphorylation of serine 29 inhibits the E3 ligase Stub1-mediated ubiquitination of PYCR1, thereby stabilizing its protein level. Based on our findings, it was determined that the phosphorylation of serine 29 in PYCR1 by ITPKA enhances the stability of the phosphorylated PYCR1 protein. This, in turn, involved significantly in oncogenic function of ITPKA in glioblastoma. Consequently, ITPKA holds promise as a potential target in prospective glioma therapy.

2.
Virol J ; 21(1): 109, 2024 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-38734674

RESUMO

BACKGROUND: Syndrome coronavirus-2 (SARS-CoV-2) has developed various strategies to evade the antiviral impact of type I IFN. Non-structural proteins and auxiliary proteins have been extensively researched on their role in immune escape. Nevertheless, the detailed mechanisms of structural protein-induced immune evasion have not been well elucidated. METHODS: Human alveolar basal epithelial carcinoma cell line (A549) was stimulated with polyinosinic-polycytidylic acid (PIC) and independently transfected with four structural proteins expression plasmids, including nucleocapsid (N), spike (S), membrane (M) and envelope (E) proteins. By RT-qPCR and ELISA, the structural protein with the most pronounced inhibitory effects on IFN-ß induction was screened. RNA-sequencing (RNA-Seq) and two differential analysis strategies were used to obtain differentially expressed genes associated with N protein inhibition of IFN-ß induction. Based on DIANA-LncBase and StarBase databases, the interactive competitive endogenous RNA (ceRNA) network for N protein-associated genes was constructed. By combining single-cell sequencing data (GSE158055), lncRNA-miRNA-mRNA axis was further determined. Finally, RT-qPCR was utilized to illustrate the regulatory functions among components of the ceRNA axis. RESULTS: SARS-CoV-2 N protein inhibited IFN-ß induction in human alveolar epithelial cells most significantly compared with other structural proteins. RNA-Seq data analysis revealed genes related to N protein inhibiting IFNs induction. The obtained 858 differentially expressed genes formed the reliable ceRNA network. The function of LINC01002-miR-4324-FRMD8 axis in the IFN-dominated immune evasion was further demonstrated through integrating single-cell sequencing data. Moreover, we validated that N protein could reverse the effect of PIC on LINC01002, FRMD8 and miR-4324 expression, and subsequently on IFN-ß expression level. And LINC01002 could regulate the production of FRMD8 by inhibiting miR-4324. CONCLUSION: SARS-CoV-2 N protein suppressed the induction of IFN-ß by regulating LINC01002 which was as a ceRNA, sponging miR-4324 and participating in the regulation of FRMD8 mRNA. Our discovery provides new insights into early intervention therapy and drug development on SARS-CoV-2 infection.


Assuntos
COVID-19 , MicroRNAs , RNA Longo não Codificante , Humanos , Células A549 , Proteínas do Nucleocapsídeo de Coronavírus/genética , Proteínas do Nucleocapsídeo de Coronavírus/metabolismo , COVID-19/virologia , COVID-19/imunologia , Evasão da Resposta Imune , Interferon beta/genética , Interferon beta/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Fosfoproteínas , RNA Endógeno Competitivo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
3.
BMC Cancer ; 23(1): 1159, 2023 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-38017386

RESUMO

BACKGROUND: As a histone methyltransferase, suppressor of variegation 3-9 homolog 1 (SUV39H1) plays an important role in the occurrence and development of cancer. To explore the mechanism and biological function of SUV39H1 in hepatitis B virus-associated hepatocellular carcinoma (HBV-HCC) can gain an insight into the pathogenesis of HBV-HCC. METHODS: The effect of HBV infection on SUV39H1 in hepatoma cells was detected. CCK-8, colony growth assay and wound healing assay were used to assess the proliferation and migration of HBV-positive hepatoma cells. RNA sequencing (RNA-seq) was applied to find differential genes and enriched pathways. The serum SUV39H1 level in HBV-HCC patients was detected and its correlation with clinical indicators was analyzed. RESULTS: SUV39H1 was increased by HBV infection and promoted the proliferation and migration of hepatoma cells. SUV39H1 could upregulate the expression of mitochondrial oxidative phosphorylation (OXPHOS) pathway-related genes. OXPHOS pathway inhibitors could reduce the capacity of proliferation and migration of hepatoma cells after overexpressing SUV39H1. Serum SUV39H1 levels were higher in chronic hepatitis B (CHB) patients than in healthy controls and higher in HBV-HCC patients than in CHB patients. In the diagnosis of HCC, the predictive value of SUV39H1 combined with alpha-fetoprotein (AFP) was better than that of AFP alone. CONCLUSION: SUV39H1 is regulated by HBV infection and promotes the proliferation and migration of hepatoma cells by targeting OXPHOS pathway. It indicates that SUV39H1 may be a new biomarker of the diagnosis of HCC.


Assuntos
Carcinoma Hepatocelular , Hepatite B Crônica , Hepatite B , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patologia , Vírus da Hepatite B/metabolismo , alfa-Fetoproteínas/metabolismo , Neoplasias Hepáticas/patologia , Fosforilação Oxidativa , Biomarcadores , Hepatite B/complicações , Hepatite B Crônica/complicações , Hepatite B Crônica/patologia , Metiltransferases/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
4.
Front Immunol ; 14: 1222425, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37662915

RESUMO

Cell migration-inducing protein (CEMIP), also known as KIAA1199 and hyaluronan-binding protein involved in hyaluronan depolymerization, is a new member of the hyaluronidase family that degrades hyaluronic acid (HA) and remodels the extracellular matrix. In recent years, some studies have reported that CEMIP can promote the proliferation, invasion, and adhesion of various tumor cells and can play an important role in bacterial infection and arthritis. This review focuses on the pathological mechanism of CEMIP in a variety of diseases and expounds the function of CEMIP from the aspects of inhibiting cell apoptosis, promoting HA degradation, inducing inflammatory responses and related phosphorylation, adjusting cellular microenvironment, and regulating tissue fibrosis. The diagnosis and treatment strategies targeting CEMIP are also summarized. The various functions of CEMIP show its great potential application value.


Assuntos
Artrite , Ácido Hialurônico , Humanos , Hialuronoglucosaminidase , Apoptose , Movimento Celular
5.
Arterioscler Thromb Vasc Biol ; 43(6): 1015-1030, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37051931

RESUMO

BACKGROUND: AGK (acylglycerol kinase) was first identified as a mitochondrial transmembrane protein that exhibits a lipid kinase function. Recent studies have established that AGK promotes cancer growth and metastasis, enhances glycolytic metabolism and function fitness of CD8+ T cells, or regulates megakaryocyte differentiation. However, the role of AGK in platelet activation and arterial thrombosis remains to be elaborated. METHODS: We performed hematologic analysis using automated hematology analyzer and investigated platelets morphology by transmission electron microscope. We explored the role of AGK in platelet activation and arterial thrombosis utilizing transgenic mice, platelet functional experiments in vitro, and thrombosis models in vivo. We revealed the regulation effect of AGK on Talin-1 by coimmunoprecipitation, mass spectrometry, immunofluorescence, and Western blot. We tested the role of AGK on lipid synthesis of phosphatidic acid/lysophosphatidic acid and thrombin generation by specific Elisa kits. RESULTS: In this study, we found that AGK depletion or AGK mutation had no effect on the platelet average volumes, the platelet microstructures, or the expression levels of the major platelet membrane receptors. However, AGK deficiency or AGK mutation conspicuously decreased multiple aspects of platelet activation, including agonists-induced platelet aggregation, granules secretion, JON/A binding, spreading on Fg (fibrinogen), and clot retraction. AGK deficiency or AGK mutation also obviously delayed arterial thrombus formation but had no effect on tail bleeding time and platelet procoagulant function. Mechanistic investigation revealed that AGK may promote Talin-1Ser425 phosphorylation and affect the αIIbß3-mediated bidirectional signaling pathway. However, AGK does not affect lipid synthesis of phosphatidic acid/lysophosphatidic acid in platelets. CONCLUSIONS: AGK, through its kinase activity, potentiates platelet activation and arterial thrombosis by promoting Talin-1 Ser425 phosphorylation and affecting the αIIbß3-mediated bidirectional signaling pathway.


Assuntos
Talina , Trombose , Animais , Camundongos , Plaquetas/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Camundongos Transgênicos , Ácidos Fosfatídicos/metabolismo , Ácidos Fosfatídicos/farmacologia , Ativação Plaquetária , Agregação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Transdução de Sinais , Talina/genética , Talina/metabolismo , Talina/farmacologia , Trombose/patologia
6.
Thromb Haemost ; 123(3): 295-306, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36402131

RESUMO

The adhesion G protein-coupled receptor GPR56 mediates cell-cell and cell-extracellular matrix interactions. To examine the function of GPR56 in platelet activation and arterial thrombosis, we generated GPR56-knockout mice and evaluated GPR56 expression in human and mouse platelets. The results revealed that the levels of the GPR56 N-terminal fragment were significantly higher on the first day after myocardial infarction than on the seventh day in the plasma of patients with ST-segment-elevation myocardial infarction. Next, we investigated the effects of GPR56 on platelet function in vitro and in vivo. We observed that collagen-induced aggregation and adenosine triphosphate release were reduced in Gpr56 -/- platelets. Furthermore, P-selectin expression on the Gpr56 -/- platelet surface was also reduced, and the spreading area on immobilized collagen was decreased in Gpr56 -/- platelets. Furthermore, collagen-induced platelet activation in human platelets was inhibited by an anti-GPR56 antibody. Gpr56 -/- mice showed an extended time to the first occlusion in models with cremaster arteriole laser injury and FeCl3-induced carotid artery injury. GPR56 activated the G protein 13 signaling pathway following collagen stimulation, which promoted platelet adhesion and thrombus formation at the site of vascular injury. Thus, our study confirmed that GPR56 regulated the formation of arterial thrombosis. Inhibition of the initial response of GPR56 to collagen could significantly inhibit platelet activation and thrombus formation. Our results provide new insights for research into antiplatelet drugs.


Assuntos
Infarto do Miocárdio , Trombose , Humanos , Camundongos , Animais , Agregação Plaquetária , Ativação Plaquetária , Plaquetas/metabolismo , Camundongos Knockout , Colágeno/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
7.
Front Microbiol ; 13: 1052917, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36504808

RESUMO

Background: Chronic hepatitis B (CHB) remains a significant global health problem, leading to recurrent inflammation and liver-damaging diseases such as fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). Currently, although diagnostic markers for CHB are well established, the indicators for predicting liver injury caused by hepatitis B virus (HBV) infection still need to be further explored. Thus, the identification of credible infectious indicators is urgently needed to facilitate timely clinical intervention and avoid the progression of disease malignancy. Methods: The Gene Expression Omnibus (GEO) database GSE83148 data set was used to explore the hub genes for HBV infection. The quantitative real-time polymerase chain reaction (qPCR) was used to identify the impact of HBV infection on the expression of hub gene at the cell level. At the same time, serum samples and clinical information were collected from healthy, HBV-free and CHB patients. The enzyme-linked immunosorbent assay (ELISA) was used to verify the results of cell experiments and Pearson correlation analysis was used to clarify hub genes correlation with HBV infection indicators and liver injury-related indicators. Finally, the Gene Expression Profiling Interactive Analysis (GEPIA) database was used to analyze the differences in the expression of hub gene in liver injury diseases. Results: Chemokine (C-X-C motif) ligand (CXCL)8, CXCL9, CXCL10, and CXCL11 were identified as hub genes in HBV infection. After HBV infection, the expression of the four chemokines was significantly increased and the concentrations secreted into serum were also increased. Moreover, the four chemokines were significantly correlated with HBV infection-related indicators and liver injury-related indicators, which were positively correlated with alanine aminotransferase (ALT), aspartate aminotransferase (AST) and hepatitis B e antigen (HBeAg), and negatively correlated with AST/ALT ratio and hepatitis B core antibody (HBcAb). In addition, the expression of CXCL9, CXCL10, and CXCL11 in HCC tissues was significantly higher than in normal tissues. Conclusion: Using a combination of bioinformatics, cell experiments, and clinical correlation analysis, this study showed that CXCL8, CXCL9, CXCL10, and CXCL11 can be used as serum biomarkers to forecast liver injury caused by HBV infection.

8.
Front Immunol ; 13: 875786, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35898493

RESUMO

Background: JMJD8 has recently been identified as a cancer-related gene, but current studies provide limited information. We aimed to clarify its roles and the potential mechanisms in pan-cancer. Methods: Pan-cancer bulk sequencing data and online web tools were applied to analyze JMJD8's correlations with prognosis, genome instability, cancer stemness, DNA repair, and immune infiltration. Moreover, single-cell datasets, SpatialDB database, and multiple fluorescence staining were used to validate the association between JMJD8 expression and M2 macrophages. Further, we utilized ROCplotter and cMap web tool to analyze the therapeutic responses and screened JMJD8-targeted compounds, respectively, and we used AlphaFold2 and Discovery Studio to conduct JMJD8 homology modeling and molecular docking. Results: We first noticed that JMJD8 was an oncogene in many cancer types. High JMJD8 was associated with lower genome stability. We then found that high JMJD8 correlated with high expression of mismatch repair genes, stemness, homologous repair gene signature in more than 9 cancers. ESTIMATE and cytokine analyses results presented JMJD8's association with immunosuppression. Also, immune checkpoint CD276 was positively relevant to JMJD8. Subsequently, we validated JMJD8 as the M2 macrophage marker and showed its connection with other immunosuppressive cells and CD8+ T-cell depression. Finally, potential JMJD8-targeted drugs were screened out and docked to JMJD8 protein. Conclusion: We found that JMJD8 was a novel oncogene, and it correlated with immunosuppression and DNA repair. JMJD8 was highly associated with immune checkpoint CD276 and was an M2 macrophage biomarker in many cancers. This study will reveal JMJD8's roles in pan-cancer and its potential as a novel therapeutic target.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Histona Desmetilases com o Domínio Jumonji/metabolismo , Neoplasias , Antígenos B7/genética , Biomarcadores , Dano ao DNA , Reparo do DNA/genética , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Terapia de Imunossupressão , Macrófagos , Simulação de Acoplamento Molecular , Neoplasias/tratamento farmacológico , Neoplasias/genética
9.
Exp Cell Res ; 416(1): 113136, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35421367

RESUMO

Glioma is one of the most common malignancies. De novo serine synthesis promotes glioma progression and therapeutic resistance. Therefore, clarifying the regulatory mechanism of serine synthesis is of great significance for glioma therapy. In this study, we found that the expression of TFCP2 was upregulated in glioma and that TFCP2 promoted glioma cell growth and sphere formation. Knockdown of TFCP2 expression inhibited glioma cell growth, sphere formation and tumorigenicity in nude mice. In terms of its molecular mechanism, TFCP2 was found to interact with ATF3 to cooperatively regulate the de novo synthesis of serine. Knockdown of TFCP2 expression significantly inhibited the binding of ATF3 to the promoter of PHGDH (a rate-limiting enzyme in the serine synthesis process). In conclusion, our studies proved that TFCP2 jointly regulates the de novo synthesis of serine through interaction with ATF3, thus promoting glioma progression. This study suggests that TFCP2 is a potential target for glioma therapy.


Assuntos
Glioma , Serina , Animais , Proteínas de Transporte , Linhagem Celular Tumoral , Proteínas de Ligação a DNA , Glioma/genética , Camundongos , Camundongos Nus , Serina/metabolismo , Fatores de Transcrição/metabolismo
10.
Aging (Albany NY) ; 13(18): 22109-22119, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34536269

RESUMO

PURPOSE: LHX9 methylation has been reported in many tumors, but its functions and related mechanisms in glioma are still unknown and need to be verified. METHODS: The protein level of LHX9 in glioma tissues was examined using western blotting and immunohistochemistry, and the functions of LHX9 in glioma cell lines were investigated using MTT and colony formation assays. In addition, the interaction between LHX9 and P53 was analyzed by immunoprecipitation, and the roles of LHX9 in cancer metabolism were explored by measuring metabolites. RESULTS: In this study, we found that the LHX9 expression level was decreased in glioma specimens, and the upregulation of LHX9 expression inhibited the growth of glioma cells in liquid medium and on soft agar. Regarding the molecular mechanism, we found that LHX9 interacted with p53, and downregulation of LHX9 promoted the expression of the glycolysis-related enzyme PGK1 and increased the lactic acid content. By interfering with the expression of LHX9, the tumorigenicity of glioma cells was promoted, an outcome blocked by further interference with PGK1 expression. CONCLUSION: In summary, the decreased expression of LHX9 in gliomas activates the expression of the glycolysis-related enzyme PGK1, thereby promoting the development of gliomas, suggesting that the LHX9-PGK1 signaling axis can be used as a target for the treatment of glioma.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Glicólise , Proteínas com Homeodomínio LIM/metabolismo , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/fisiopatologia , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Glioma/fisiopatologia , Humanos , Proteínas com Homeodomínio LIM/genética , Masculino , Camundongos , Camundongos Nus , Ligação Proteica , Transdução de Sinais , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/genética
11.
Chin J Traumatol ; 24(5): 273-279, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34016503

RESUMO

PURPOSE: Low-velocity penetrating brain injury (LVPBI) caused by foreign bodies can pose life-threatening emergencies. Their complexity and lack of validated classification data have prevented standardization of clinical management. We aimed to compare the trans-base and trans-vault phenotypes of LVPBI to help provide guidance for clinical decision-making of such injury type. METHODS: A retrospective study on LVPBI patients managed at our institution from November 2013 to March 2020 was conducted. We included LVPBI patients admitted for the first time for surgery, and excluded those with multiple injuries, gunshot wounds, pregnancy, severe blunt head trauma, etc. Patients were categorized into trans-base and trans-vault LVPBI groups based on the penetration pathway. Discharged patients were followed up by outpatient visit or telephone. The data were entered into the Electronic Medical Record system by clinicians, and subsequently derived by researchers. The demography and injury characteristics, treatment protocols, complications, and outcomes were analyzed and compared between the two groups. A t-test was used for analysis of normally distributed data, and a Mann-Whitney U test for non-parametric data. A generalized linear model was further established to determine whether the factors length of stay and performance scale score were influenced by each factor. RESULTS: A total of 27 LVPBI patients were included in this analysis, comprised of 13 (48.1%) trans-base cases and 14 (51.9%) trans-vault cases. Statistical analyses suggested that trans-base LVPBI was correlated with deeper wounds; while the trans-vault phenotype was correlated with injury by metal foreign bodies. There was no difference in Glasgow Coma Scale score and the risk of intracranial hemorrhage between the two groups. Surgical approaches in the trans-base LVPBI group included subfrontal (n = 5, 38.5%), subtemporal (n = 5, 38.5%), lateral fissure (n = 2, 15.4%), and distal lateral (n = 1, 7.7%). All patients in the trans-vault group underwent a brain convex approach using the foreign body as reference (n = 14, 100%). Moreover, the two groups differed in application prerequisites for intracranial pressure monitoring and vessel-related treatment. Trans-base LVPBI was associated with higher rates of cranial nerve and major vessel injuries; in contrast, trans-vault LVPBI was associated with lower functional outcome scores. CONCLUSION: Our findings suggest that trans-base and trans-vault LVPBIs differ in terms of characteristics, treatment, and outcomes. Further understanding of these differences may help guide clinical decisions and contribute to a better management of LVPBIs.


Assuntos
Traumatismos Cranianos Penetrantes , Ferimentos por Arma de Fogo , Escala de Coma de Glasgow , Traumatismos Cranianos Penetrantes/diagnóstico por imagem , Traumatismos Cranianos Penetrantes/cirurgia , Humanos , Prognóstico , Estudos Retrospectivos
12.
Biosci Rep ; 40(10)2020 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-33057597

RESUMO

Glioblastoma multiforme (GBM) is one of the most malign brain tumors in adults. Temozolomide (TMZ) is an oral chemotherapy drug constituting the backbone of chemotherapy regimens utilized as first-line treatment of GBM. However, resistance to TMZ often leads to treatment failure. In the present study, we explored the expression and related mechanisms of nuclear enriched abundant transcript 1 (NEAT1) in glioma stem cells (GSCs). Quantitative real-time PCR (qRT-PCR) showed that NEAT1 was up-regulated in serum samples of GBM patients and GSCs isolated from U87, U251 cell lines. Functional experiments showed that NEAT1 knockdown restrained malignant behaviors of GSC, including proliferation, migration and invasion. Dual-luciferase assays identified let-7g-5p was a downstream target and negatively adjusted by NEAT1. Restoration of let-7g-5p impeded tumor progression by inhibiting proliferation, migration and invasion. Mitogen-activated protein kinase kinase kinase 1 (MAP3K1), as a direct target of let-7g-5p, was positively regulated by NEAT1 and involved to affect the regulation of NEAT1 on GSCs' behaviors. In conclusion, our results suggested that NEAT1 promoted GSCs progression via NEAT1/let-7g-5p/MAP3K1 axis, which provided a depth insight into TMZ resistance mechanism.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/genética , Glioblastoma/tratamento farmacológico , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Temozolomida/farmacologia , Neoplasias Encefálicas/sangue , Neoplasias Encefálicas/genética , Estudos de Casos e Controles , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Técnicas de Silenciamento de Genes , Glioblastoma/sangue , Glioblastoma/genética , Voluntários Saudáveis , Humanos , MAP Quinase Quinase Quinase 1/genética , Invasividade Neoplásica/genética , Células-Tronco Neoplásicas/patologia , RNA Longo não Codificante/sangue , Temozolomida/uso terapêutico , Regulação para Cima
13.
FASEB J ; 34(6): 7360-7371, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32350920

RESUMO

It has been documented that M2 macrophage polarization plays a suppressive role in atherosclerosis in diabetes mellitus (DM). In addition, prostaglandin E2 (PGE2) is implicated in the development of M2 macrophage polarization. Therefore, the study aimed to investigate the specific mechanism of PGE2 in M2 macrophage polarization in diabetic coronary atherosclerosis (DMAS). Initially, clinical samples were obtained and DMAS mouse model was established. The expression of BDNF was determined, and M1 and M2 macrophage polarizations were evaluated. Then, the levels of BDNF and PGE2 were modified in DMAS mice and the serum indicator, atherosclerotic plaque, lipid uptake by PBMCs, as well as M1 and M2 macrophage polarization were determined. Macrophages were isolated and the effects of PGE2 and the CREB/BDNF/TrkB signaling pathway on M2 macrophage polarization were explored. BDNF was downregulated and macrophages were differentiated into M1 in DMAS patients and mice. BDNF and PGE2 were observed to promote M2 macrophage polarization, where atherosclerotic plaque and lipid uptake by PBMCs were reduced, and DMAS was alleviated in mice. Overexpression of BDNF activated the CREB/BDNF/TrkB signaling pathway and stimulated M2 macrophage polarization in macrophages. PGE2 stimulated M2 macrophage polarization by inducing KLF4 via the activation of the CREB/BDNF/TrkB signaling pathway. This study demonstrates that PGE2 promotes M2 macrophage polarization by activating the CREB/BDNF/TrkB signaling pathway, thus alleviating DMAS.


Assuntos
Aterosclerose/tratamento farmacológico , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Polaridade Celular/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Dinoprostona/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Glicoproteínas de Membrana/metabolismo , Receptor trkB/metabolismo , Animais , Aterosclerose/metabolismo , Diferenciação Celular/efeitos dos fármacos , Doença da Artéria Coronariana/tratamento farmacológico , Doença da Artéria Coronariana/metabolismo , Diabetes Mellitus/metabolismo , Feminino , Humanos , Fator 4 Semelhante a Kruppel , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Placa Aterosclerótica/tratamento farmacológico , Placa Aterosclerótica/metabolismo , Células RAW 264.7 , Transdução de Sinais/efeitos dos fármacos
14.
Anal Chim Acta ; 1109: 130-139, 2020 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-32252896

RESUMO

A novel three-dimensional nickel hydroxide/polyurethane (Ni(OH)2/PU) electrode was prepared by a simple and environmentally friendly method and used for non-enzymatic detection of glucose. The Ni(OH)2/PU electrode was obtained by one-pot hydrothermal method of loading nickel hydroxide on a cheap, easily available and flexible polyurethane sponge, which is facile and energy-saving. The porous structure of the polyurethane sponge provides a large surface area and a rich electrochemical active site for the electrode, which is beneficial to the oxidation reaction of glucose on the surface of the electrode with Ni(OH)2. The Ni(OH)2/PU electrode structure was characterized by scanning electron microscopy (SEM), transmission electron microscopy (TEM), X-ray diffraction (XRD) and X-ray photoelectron spectroscopy (XPS). The cyclic voltammetry test was used to study the catalytic performance of Ni(OH)2/PU electrode for oxidation of glucose and the chronoamperometry was used to investigate the detection performance of Ni(OH)2/PU electrode on glucose. The results indicate that this non-enzymatic glucose sensor had a high sensitivity of 2845 µA mM-1 cm-2, a low detection limit of 0.32 µM (S/N = 3), a detection range of 0.01-2.06 mM and response time of less than 5 s. In addition, the Ni(OH)2/PU electrode had excellent selectivity, reproducibility and stability and also exhibited effective detection of glucose in fetal bovine serum (FBS). In summary, Ni(OH)2/PU electrode had broad prospects as an excellent candidate for non-enzymatic glucose sensors. The study also opens up a facile and energy-saving approach for preparing three-dimensional (3D) functionalized polymer electrode via hydrothermal method as electrochemical sensors.


Assuntos
Técnicas Biossensoriais , Técnicas Eletroquímicas , Glucose/análise , Hidróxidos/química , Níquel/química , Poliuretanos/química , Eletrodos
15.
Cell Signal ; 70: 109569, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32061924

RESUMO

Diabetes mellitus-accelerated atherosclerosis (DMAS) is one of the vascular complications of diabetes. Brain-derived neurotrophic factor (BDNF) plays a critical role in diabetes mellitus. However, the mechanism by which BDNF is involved in DMAS remains unknown. This study investigates the effect of BDNF on the progression of DMAS as well as the underlying mechanism of action. The levels of BDNF in serum and peripheral blood mononuclear cells (PBMCs) from patients with DMAS and health controls were measured as well as the expression of inflammatory cytokines (IL-1ß, TNF-α, IL-10, TGF-ß and IL-13). The effects of BDNF restoration on cytokine release, macrophage differentiation and the formation of atherosclerotic plaques were evaluated both in vitro and in vivo using the DMAS mouse model. Downregulation of BDNF was identified in the serum and PBMCs of patients with DMAS. Elevation of BDNF contributed to a reduction in the AS lesion area in low-density lipoprotein receptor-/- mice, inactivated the STAT3 pathway, decreased pro-inflammatory cytokines IL-1ß and TNF-α, and increased IL-10, TGF-ß and IL-13. BDNF overexpression also increased the proportion of M2 macrophages and alleviated atherosclerotic lesions. Our findings demonstrate that BDNF overexpression promotes M2 macrophage polarization, which represses the development of DMAS by inactivating the STAT3 pathway.


Assuntos
Aterosclerose/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Diabetes Mellitus/metabolismo , Macrófagos/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Polaridade Celular , Citocinas/metabolismo , Humanos , Ativação de Macrófagos , Macrófagos/citologia , Camundongos , Camundongos Knockout
16.
Cell Stress Chaperones ; 24(1): 235-245, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30632064

RESUMO

Acute brain reperfusion stress is associated with mitochondrial dysfunction through unknown mechanisms. Accordingly, there is no effective drug to control the development and progression of brain reperfusion stress currently. The aim of our investigation is to verify whether melatonin attenuates acute brain reperfusion stress via affecting mitochondrial function. Our studies demonstrated that melatonin treatment suppressed reperfusion-induced neuron death. At the molecular levels, melatonin treatment modulated mitochondrial homeostasis via activating mitochondrial fusion. At the stage of reperfusion, MFN2 expression was downregulated, contributing to mitochondrial fusion inhibition. Interestingly, MFN2-related mitochondrial fusion was reversed by melatonin. Loss of MFN2-related mitochondrial fusion abrogated the protective actions of melatonin on mitochondrial function. Mechanistically, melatonin sustained MFN2-related mitochondrial fusion via suppressing Mst1-Hippo pathway. Overexpression of Mst1 attenuated the beneficial effects of melatonin on mitochondrial fusion, evoking mitochondrial damage and neuron death in the setting of brain reperfusion stress. Taken together, our results confirmed the protective effects of melatonin on acute brain reperfusion stress. Melatonin treatment activated MFN2-related mitochondrial fusion via suppressing Mst1-Hippo pathway, finally sustaining mitochondrial function and reducing reperfusion-mediated cerebral injury.


Assuntos
Encéfalo/patologia , GTP Fosfo-Hidrolases/metabolismo , Melatonina/uso terapêutico , Mitocôndrias/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/patologia , Transdução de Sinais , Estresse Fisiológico , Doença Aguda , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Metabolismo Energético/efeitos dos fármacos , GTP Fosfo-Hidrolases/deficiência , Técnicas de Silenciamento de Genes , Fator de Crescimento de Hepatócito/metabolismo , Via de Sinalização Hippo , Melatonina/farmacologia , Camundongos , Mitocôndrias/efeitos dos fármacos , Dinâmica Mitocondrial , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Estresse Fisiológico/efeitos dos fármacos
17.
J Cell Physiol ; 234(4): 4054-4067, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30206929

RESUMO

Papillary thyroid cancer (PTC) is a kind of thyroid cancer and frequently presents with epithelial-mesenchymal transition (EMT). MicroRNAs (miRNAs) were previously reported to be associated with PTC. Thus, this study aims to define the role of microRNA-520a-3p (miR-520a-3p) in PTC through the JAK/STAT signaling pathway by targeting JAK1. The PTC and normal thyroid tissues of 137 PTC patients were collected. First of all, the expression pattern of miR-520a-3p, JAK1, JAK2, STAT3, E-cadherin, and vimentin in PTC was identified. The relationship between miR-520a-3p and JAK1 was predicted and analyzed. And a series of miR-520a-3p mimic or inhibitor, or siRNA JAK1 introduced into PTC cells were applied to examine the effect of miR-520a-3p on PTC cell viability, migration, invasion, cell cycle, apoptosis, and EMT. Meanwhile, the regulatory effect of miR-520a-3p and JAK1 on the JAK/STAT signaling pathway was also determined. The expression of JAK1, JAK2, STAT3, and vimentin increased yet miR-520a-3p and E-cadherin decreased in PTC tissue. JAK1 was negatively regulated by miR-520a-3p. Functionally, EMT induction was prevented by miR-520a-3p upregulation through downregulating JAK1. When upregulating miR-520a-3p or silencing JAK1 in PTC cells, PTC cell viability, migration, and invasion were inhibited yet cell apoptosis promoted with cells arrested at G1 phase, indicating that miR-520a-3p prevented PTC progression by downregulating JAK1. Moreover, miR-520a-3p elevation or JAK1 inhibition inactivated the JAK/STAT signaling pathway. Collectively, miR-520a-3p prevents cancer progression through inactivating the JAK/STAT signaling pathway by downregulating JAK1 in PTC.


Assuntos
Movimento Celular , Transição Epitelial-Mesenquimal , Janus Quinase 1/metabolismo , MicroRNAs/metabolismo , Fator de Transcrição STAT3/metabolismo , Câncer Papilífero da Tireoide/enzimologia , Neoplasias da Glândula Tireoide/enzimologia , Adulto , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular , Regulação Neoplásica da Expressão Gênica , Humanos , Janus Quinase 1/genética , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Invasividade Neoplásica , Fator de Transcrição STAT3/genética , Transdução de Sinais , Câncer Papilífero da Tireoide/genética , Câncer Papilífero da Tireoide/patologia , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/patologia , Adulto Jovem
18.
Oncotarget ; 8(16): 26448-26459, 2017 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-28103573

RESUMO

Several observational studies have shown that metformin can modify the risk and survival of colorectal cancer (CRC) in patients with diabetes mellitus, although the magnitude of this relationship has not been determined. We conducted an updated systematic review and meta-analysis to analyze the association between metformin and CRC mortality and searched relevant databases up to July 2016. The primary outcome was overall survival (OS). Secondary outcomes were cancer-specific survival (CS) and disease-free survival (DFS). Summary hazard ratios (HRs) were calculated using a random-effects model. Seventeen studies enrolling 269,417 participants were eligible for inclusion. Comparing with non-metformin users in diabetic CRC patients, the summary HRs for OS in metformin users were 0.69 (95% CI, 0.61-0.77). Subgroup analyses stratified by the study characteristics and sensitivity analysis by the trim-and-fill method (adjusted HR 0.77, 95% CI, 0.67-0.87) confirmed the robustness of the results. However, significant OS benefit was noted in patients with stage II and III disease. Five studies reported the CRC prognosis for CS and three for DFS; metformin intake was significantly associated with patient CS (HR 0.75, 95% CI, 0.59-0.94), but not DFS (HR 0.38, 95% CI, 0.13-1.17). Our findings suggest that metformin intake is associated with improved survival outcomes in terms of OS and CS in CRC patients with diabetes, particular for OS in stage II and stage III patients. Further studies should be conducted to determine CRC survival between metformin use and patient specific clinical and molecular profiles.


Assuntos
Neoplasias Colorretais/complicações , Neoplasias Colorretais/mortalidade , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Metformina/uso terapêutico , Estudos de Coortes , Neoplasias Colorretais/patologia , Humanos , Prognóstico , Modelos de Riscos Proporcionais , Viés de Publicação , Pesquisa Qualitativa
19.
PLoS One ; 8(12): e82309, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24376526

RESUMO

OBJECTIVE: To explore the endocrine mechanisms of aldosterone-producing adenoma (APA) by using the microarray expression profiles of normal and APA samples. METHODS: The gene expression profile GSE8514 was downloaded from Gene Expression Omnibus database, including samples from normal adrenals (n = 5) and APAs (n = 10). The differentially expressed genes (DEGs) were identified by samr package and endocrine DEGs were obtained according to Clinical Genome Database. Then, functional enrichment analysis of screened DEGs was performed by DAVID (Database for Annotation, Visualization and Integrated Discovery). Finally, a regulatory network was constructed to screen endocrine genes related with adrenal dysfunction and pathway enrichment analysis for the constructed network was performed. RESULTS: A total of 2149 DEGs were identified including 379 up- and 1770 down-regulated genes. And 26 endocrine genes were filtered from the DEGs. Furthermore, the down-regulated DEGs are mainly related to protein kinase cascade, response to molecule of bacterial origin, response to lipopolysaccharide, cellular macromolecule catabolic process and macromolecule catabolic process, while the up-regulated DEGs are related with regulation of ion transport. The target genes of VDR (vitamin D receptor), one of the three endocrine genes differentially expressed in the regulatory network, were endocrine genes including CYP24A1 (25-hydroxyvitamin D-24-hydroxylase) and PTH (parathyroid hormone). Three pathways may be associated with APA pathogenesis including cytokine-cytokine receptor interaction, pathways in cancer and autoimmune thyroid disease. CONCLUSION: The VDR is the most significant transcription factor and related endocrine genes might play important roles in the endocrine mechanisms of APA.


Assuntos
Adenoma/genética , Neoplasias das Glândulas Suprarrenais/genética , Aldosterona/biossíntese , Receptores de Calcitriol/genética , Adulto , Análise por Conglomerados , Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica , Ontologia Genética , Redes Reguladoras de Genes , Humanos , Transcriptoma , Regulação para Cima/genética
20.
Int J Biol Sci ; 10(1): 80-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24391454

RESUMO

The aim of this study is to explore the localization of human mesenchymal stem cells from umbilical cord matrix (hMSCs-UC) and the role of these cells in the repair of foot ulcerate tissue in diabetic foot ulcers in rats. A diabetic rat model was established by administering Streptozotocin. Diabetic foot ulceration was defined as non-healing or delayed-healing of empyrosis on the dorsal hind foot after 14 weeks. hMSCs-UC were delivered through the left femoral artery. We evaluated the localization of hMSCs-UC and their role in tissue repair in diabetic foot ulcers by histological analysis, PCR, and immunohistochemical staining. A model for diabetes was established in 54 out of 60 rats (90% success rate) and 27 of these rats were treated with hMSCs-UC. The area of ulceration was significantly and progressively reduced at 7 and 14 days following treatment with hMSCs-UC. This gross observation was strongly supported by the histological changes, including newly developed blood vessels and proliferation of inflammatory cells at 3 days post-treatment, significant increase in granulation tissue at 7 days post-treatment and squamous epithelium or stratified squamous epithelium at 14 days post-treatment. Importantly, human leukocyte antigen type-I (HLA-1) was confirmed in ulcerated tissue by RT-PCR. The expression of cytokeratin 19 was significantly increased in diabetic model rats, with no detectable change in cytokeratin 10. Additionally, both collagens I and III increased in model rats treated with hMSCs-UC, but the ratio of collagen I/III was less significant in treated rats compared with control rats. These results suggest that hMSCs-UC specifically localize to the target ulcerated tissue and may promote the epithelialization of ulcerated tissue by stimulating the release of cytokeratin 19 from keratinocytes and extracellular matrix formation.


Assuntos
Pé Diabético/terapia , Sangue Fetal/citologia , Células-Tronco Mesenquimais/citologia , Cordão Umbilical/citologia , Animais , Sequência de Bases , Colágeno/metabolismo , Primers do DNA , Queratinas/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estreptozocina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA