Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Neuro Oncol ; 2024 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-38456228

RESUMO

BACKGROUND: Hypoxia is associated with poor prognosis in many cancers including glioblastoma (GBM). Glioma stem-like cells (GSCs) often reside in hypoxic regions and serve as reservoirs for disease progression. Long non-coding RNAs (lncRNAs) have been implicated in GBM. However, the lncRNAs that modulate GSC adaptations to hypoxia are poorly understood. Identification of these lncRNAs may provide new therapeutic strategies to target GSCs under hypoxia. METHODS: lncRNAs induced by hypoxia in GSCs were identified by RNAseq. LUCAT1 expression was assessed by qPCR, RNAseq, Northern blot, single molecule FISH in GSCs, and interrogated in IvyGAP, TCGA, and CGGA databases. LUCAT1 was depleted by shRNA, CRISPR/Cas9, and CRISPR/Cas13d. RNAseq, Western blot, immunohistochemistry, co-IP, ChIP, ChIPseq, RNA immunoprecipitation, and proximity ligation assay were performed to investigate mechanisms of action of LUCAT1. GSC viability, limiting dilution assay, and tumorigenic potential in orthotopic GBM xenograft models were performed to assess the functional consequences of depleting LUCAT1. RESULTS: A new isoform of Lucat1 is induced by HIF1α and NRF2 in GSCs under hypoxia. LUCAT1 is highly expressed in hypoxic regions in GBM. Mechanistically, LUCAT1 formed a complex with HIF1α and its co-activator CBP to regulate HIF1α target gene expression and GSC adaptation to hypoxia. Depletion of LUCAT1 impaired GSC self-renewal. Silencing LUCAT1 decreased tumor growth and prolonged mouse survival in GBM xenograft models. CONCLUSIONS: A HIF1α-LUCAT1 axis forms a positive feedback loop to amplify HIF1α signaling in GSCs under hypoxia. LUCAT1 promotes GSC self-renewal and GBM tumor growth. LUCAT1 is a potential therapeutic target in GBM.

2.
Cancer Sci ; 115(5): 1587-1601, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38438251

RESUMO

Both lysine and arginine methyltransferases are thought to be promising therapeutic targets for malignant tumors, yet how these methyltransferases function in malignant tumors, especially hepatocellular carcinoma (HCC), has not been fully elucidated. Here, we reported that SMYD4, a lysine methyltransferase, acts as an oncogene in HCC. SMYD4 was highly upregulated in HCC and promoted HCC cell proliferation and metastasis. Mechanistically, PRMT5, a well-known arginine methyltransferase, was identified as a SMYD4-binding protein. SMYD4 monomethylated PRMT5 and enhanced the interaction between PRMT5 and MEP50, thereby promoting the symmetrical dimethylation of H3R2 and H4R3 on the PRMT5 target gene promoter and subsequently activating DVL3 expression and inhibiting expression of E-cadherin, RBL2, and miR-29b-1-5p. Moreover, miR-29b-1-5p was found to inversely regulate SMYD4 expression in HCC cells, thus forming a positive feedback loop. Furthermore, we found that the oncogenic effect of SMYD4 could be effectively suppressed by PRMT5 inhibitor in vitro and in vivo. Clinically, high coexpression of SMYD4 and PRMT5 was associated with poor prognosis of HCC patients. In summary, our study provides a model of crosstalk between lysine and arginine methyltransferases in HCC and highlights the SMYD4-PRMT5 axis as a potential therapeutic target for the treatment of HCC.


Assuntos
Carcinoma Hepatocelular , Proliferação de Células , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas , MicroRNAs , Proteína-Arginina N-Metiltransferases , Proteína-Arginina N-Metiltransferases/metabolismo , Proteína-Arginina N-Metiltransferases/genética , Humanos , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Animais , Linhagem Celular Tumoral , MicroRNAs/genética , MicroRNAs/metabolismo , Proliferação de Células/genética , Camundongos , Metilação , Masculino , Histona-Lisina N-Metiltransferase/metabolismo , Histona-Lisina N-Metiltransferase/genética , Retroalimentação Fisiológica , Feminino , Camundongos Nus
3.
J Exp Clin Cancer Res ; 42(1): 339, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38098044

RESUMO

BACKGROUND: Within the tumor immune microenvironment (TME), tumor-associated macrophages (TAMs) are crucial in modulating polarization states to influence cancer development through metabolic reprogramming. While long non-coding RNAs (lncRNAs) have been shown to play a pivotal role in the progression of various cancers, the underlying mechanisms by which lncRNAs alter M2 polarization through macrophage metabolism remodeling remain unelucidated. METHODS: RNA sequencing was used to screen for differentially expressed lncRNAs in TAMs and normal tissue-resident macrophages (NTRMs) isolated from pancreatic ductal adenocarcinoma (PDAC) tissues, whilst RT-qPCR and FISH were employed to detect the expression level of SNHG17. Moreover, a series of in vivo and in vitro experiments were conducted to assess the functions of SNHG17 from TAMs in the polarization and glycolysis of M2-like macrophages and in the proliferation and metastasis of pancreatic cancer cells (PCs). Furthermore, Western blotting, RNA pull-down, mass spectrometry, RIP, and dual-luciferase assays were utilized to explore the underlying mechanism through which SNHG17 induces pro-tumor macrophage formation. RESULTS: SNHG17 was substantially enriched in TAMs and was positively correlated with a worse prognosis in PDAC. Meanwhile, functional assays determined that SNHG17 promoted the malignant progression of PCs by enhancing M2 macrophage polarization and anaerobic glycolysis. Mechanistically, SNHG17 could sponge miR-628-5p to release PGK1 mRNA and concurrently interact with the PGK1 protein, activating the pro-tumorigenic function of PGK1 by enhancing phosphorylation at the T168A site of PGK1 through ERK1/2 recruitment. Lastly, SNHG17 knockdown could reverse the polarization status of macrophages in PDAC. CONCLUSIONS: The present study illustrated the essential role of SNHG17 and its molecular mechanism in TAMs derived from PDAC, indicating that SNHG17 might be a viable target for PDAC immunotherapy.


Assuntos
Carcinoma Ductal Pancreático , MicroRNAs , Neoplasias Pancreáticas , RNA Longo não Codificante , Humanos , Fosforilação , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Anaerobiose , Linhagem Celular Tumoral , Proliferação de Células/genética , Neoplasias Pancreáticas/patologia , Carcinoma Ductal Pancreático/patologia , Macrófagos/metabolismo , Glicólise , MicroRNAs/genética , Microambiente Tumoral , Fosfoglicerato Quinase/genética , Fosfoglicerato Quinase/metabolismo
4.
Eur J Pharmacol ; 957: 176031, 2023 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-37660967

RESUMO

Myocardial ischemia-reperfusion (I/R) injury triggers several cell death types, including apoptosis, autophagy, and ferroptosis. Licochalcone A (LCA), a natural flavonoid compound isolated from the root of Glycyrrhiza glabra, has been demonstrated to exert potential pharmacological benefits, such as antioxidant, antitumor, and anti-inflammatory activities. The present study aimed to investigate the involvement of ferroptosis in the pathogenesis of I/R and determine whether LCA can inhibit ferroptosis to prevent the myocardial I/R injury in rats. The effects of LCA on myocardial I/R injury were detected by examining the left ventricular-developed pressure and triphenyltetrazolium chloride staining. We conducted Western blotting analyses, ELISA assay, and quantitative real-time PCR to determine the levels of ferroptosis-related molecules. To demonstrate the cardioprotective effect of LCA in vitro, H9c2 and primary neonatal rat cardiomyocytes were co-treated with ferroptosis inducers (erastin, RSL3, or Fe-SP) and LCA for 16 and 24 h. Our ex vivo study showed that LCA increased the cardiac contractility, and reduced the infarct volume and ferroptosis-related biomarkers in rat hearts after I/R. Moreover, LCA reduced the levels of ferroptosis inducers-induced reactive oxygen species generation, lipid peroxidation, and ferroptosis-related biomarkers in cultured H9c2 cells and cardiomyocytes. LCA also reduced the Fe-SP-increased nuclear factor erythroid 2-related factor 2 and heme oxygenase-1 protein levels in cultured cardiomyocytes. In the present study, we showed that the LCA-induced cardioprotective effects in attenuating the myocardial I/R injury were correlated with ferroptosis regulation, and provided a possible new therapeutic strategy for prevention or therapy of the myocardial I/R injury.


Assuntos
Chalconas , Ferroptose , Animais , Ratos , Chalconas/farmacologia , Chalconas/uso terapêutico , Fenômenos Fisiológicos Cardiovasculares , Isquemia
5.
J Clin Med ; 12(13)2023 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-37445227

RESUMO

A high malondialdehyde-oxidized low-density lipoprotein (MDA-oxLDL) level is associated with atherosclerotic cardiovascular diseases and major adverse cardiovascular events. A higher cardio-ankle vascular index (CAVI) is independently associated with an increased risk of cardiovascular events, cardiovascular mortality, myocardial infarction, and stroke in patients with cardiovascular risk. Thus, this study aimed to evaluate the relationship between serum MDA-oxLDL levels and CAVI in patients with triple-vessel coronary artery disease who underwent coronary artery bypass graft (CABG) surgery. Fasting blood samples and baseline characteristics were obtained from 88 patients who had undergone CABG. A commercialized enzyme-linked immunosorbent assay was used to measure MDA-oxLDL levels. An automatic pulse wave analyzer was used to measure CAVI values, and each side of CAVI values of ≥9 was designated as arterial stiffness. In total, 47 participants were assigned to the arterial stiffness group. More patients had diabetes mellitus, were older, and had higher serum MDA-oxLDL levels in the arterial stiffness group than in the control group. A multivariate logistic regression analysis disclosed that MDA-oxLDL and diabetes mellitus were independent predictors of arterial stiffness. Moreover, according to the Spearman's correlation analysis, the serum MDA-oxLDL level was positively associated with both left and right CAVI. Serum MDA-oxLDL levels were positively associated with arterial stiffness in patients who had undergone CABG.

7.
J Cell Physiol ; 238(1): 242-256, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36538623

RESUMO

Myocardial hypertrophy is associated with a significant increase in intracellular Ca2+ , which can be induced by long-chain fatty acid. Palmitic acid methyl ester (PAME), a fatty acid ester released from adipose tissue, superior cervical ganglion, and retina, has been found to have anti-inflammation, antifibrosis, and peripheral vasodilation effects. However, the effects of PAME on cardiomyocytes are still unclear. The aim of this study was to determine whether PAME could disrupt the intracellular Ca2+ balance, leading to cardiomyocyte hypertrophy. Neonatal rat cardiomyocytes were treated with various concentrations (10-100 µM) of PAME for 1-4 days. Cytosolic Ca2+ and mitochondrial Ca2+ concentrations were examined using Fura-2 AM and Rhod-2, respectively. After treatment with PAME for 4 days, mitochondrial Ca2+ , an indicator of the state of mitochondrial permeability transition pore (MPTP), and cell death were monitored by flow cytometric analysis. ATP levels were detected using the ATP assay kit. Cardiomyocyte hypertrophy was analyzed by measuring the cardiac hypertrophy biomarker and cell area using quantitative real time-polymerase chain reaction, Western Blot analysis and immunofluorescence analysis. Our results show that PAME concentration- and time-dependently increased cytosolic and mitochondria Ca2+ through the mitochondrial calcium uniporter. Moreover, treatment with PAME for 4 days caused MPTP opening, thereby reducing ATP production and enhancing reactive oxygen species (ROS) generation, and finally led to cardiomyocyte hypertrophy. These effects caused by PAME treatment were attenuated by the G-protein coupled receptor 40 (GPR40) inhibitor. In conclusion, PAME impaired mitochondrial function, which in turn led to cardiomyocyte hypertrophy through increasing the mitochondrial Ca2+ levels mediated by activating the GPR40 signaling pathway.


Assuntos
Cálcio , Mitocôndrias , Palmitatos , Receptores Acoplados a Proteínas G , Animais , Ratos , Trifosfato de Adenosina/metabolismo , Cálcio/metabolismo , Cardiomegalia/induzido quimicamente , Cardiomegalia/metabolismo , Mitocôndrias/metabolismo , Miócitos Cardíacos/metabolismo , Palmitatos/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Células Cultivadas
8.
Int J Cardiol ; 375: 74-86, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36513286

RESUMO

BACKGROUND: Ischemia/reperfusion (I/R) is associated with severe cellular damage and death. Ferroptosis, a new form of regulated cell death caused by the accumulation of iron-mediated lipid peroxidation, has been found in several diseases including I/R injury, which was reported to be suppressed by flavonoids. Baicalein (BAI) and luteolin (Lut) are flavonoids and were shown to reduce the myocardial I/R injury. BAI was found to suppress ferroptosis in cancer cells via reducing reactive oxygen species (ROS) generation. However, the anti-ferroptosis effect of Lut on ferroptosis has not been reported. This study aimed to investigate whether ferroptosis reduction contributes to the BAI- and Lut-protected cardiomyocytes. METHODS: This research used erastin, RSL3, and Fe-SP to induce ferroptosis. Cell viability was examined using MTT assay. Annexin V-FITC, CM-H2DCFDA, and Phen Green SK diacetate (PGSK) fluorescent intensity were detected to analyze apoptotsis, ROS levels, and Fe2+ concentrations, respectively. qPCR and Western blot analysis were conducted to detect the levels of mRNA and protein, respectively. RESULTS: Our data show that BAI and Lut protected cardiomyocytes against ferroptosis caused by ferroptosis inducers and I/R. Moreover, both BAI and Lut decreased ROS and malondialdehyde (MDA) generation and the protein levels of ferroptosis markers, and restored Glutathione peroxidase 4 (GPX4) protein levels in cardiomyocytes reduced by ferroptosis inducers. BAI and Lut reduced the I/R-induced myocardium infarction and decreased the levels of Acsl4 and Ptgs2 mRNA. CONCLUSIONS: BAI and Lut could protect the cardiomyocytes against the I/R-induced ferroptosis via suppressing accumulation of ROS and MDA.


Assuntos
Traumatismo por Reperfusão Miocárdica , Miócitos Cardíacos , Ratos , Animais , Miócitos Cardíacos/metabolismo , Luteolina/farmacologia , Luteolina/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/metabolismo , RNA Mensageiro/metabolismo , Reperfusão , Isquemia/metabolismo
9.
Mol Ther Nucleic Acids ; 29: 788-802, 2022 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-36159591

RESUMO

Hepatitis B virus X protein (HBx) is considered as an oncogene in tumorigenesis and progression of hepatocellular carcinoma (HCC). In recent years, the important role of circular RNAs (circRNAs) in HCC has been increasingly demonstrated. However, the regulatory mechanisms of HBx on circRNAs remains largely unknown. In this study, we identified that a novel circRNA, circSFMBT2, was markedly downregulated by HBx. Low expression of circSFMBT2 was correlated with poor prognosis and vascular invasion. Functionally, overexpression of circSFMBT2 significantly inhibited HCC metastasis both in vitro and in vivo. The mechanism of circSFMBT2 was to as a sponge of miR-665, which is a negative regulator of tissue inhibitor of metalloproteinases 3 (TIMP3). However, HBx downregulated circSFMBT2 via the interaction with DExH-box helicase 9 (DHX9), which binds to flanking circRNA-forming introns. In conclusion, circSFMBT2, which is downregulated by HBx, acts as a tumor suppressor to inhibit tumor metastasis through the miR-665/TIMP3 axis. Our study suggests that circSFMBT2 could be a potential prognostic biomarker and therapeutic target for HCC.

10.
J Cardiovasc Dev Dis ; 9(4)2022 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-35448081

RESUMO

Old age has been proven to be related to progressed arterial or aortic stiffness. Aortic stiffness is an independent predictor of all-cause and cardiovascular disease mortalities in patients who have undergone coronary artery bypass grafting (CABG) surgery. Higher serum concentrations of adipocyte fatty-acid-binding protein (A-FABP) could be considered a predictor of aortic stiffness in patients with hypertension or diabetes mellitus. This study aims to investigate the relationships between A-FABP and aortic stiffness in patients who have received CABG. A total of 84 CABG patients were enrolled in our study from September 2018 to May 2019. Serum A-FABP levels were determined using a commercial enzyme immunoassay. Carotid−femoral pulse wave velocity (cfPWV) > 10 m/s was defined as aortic stiffness. Of the 84 CABG patients, 28 (33.3%) with aortic stiffness had a higher average age; exhibited higher rates of diabetes; and had higher serum creatinine, C-reactive protein, and A-FABP levels compared to controls. Multivariable logistic regression revealed that serum A-FABP levels (odds ratio (OR) = 1.068, 95% confidence interval (CI) 1.017−1.121, p = 0.008) and age (OR = 1.204, 95% CI 1.067−1.359, p = 0.003) were independent predictors of aortic stiffness. Multivariable stepwise linear regression revealed significant positive correlations of age and A-FABP levels with cfPWV values. Serum A-FABP level is positively correlated with cfPWV values, and a high serum A-FABP level is associated with aortic stiffness in patients who have undergone CABG.

11.
J Exp Clin Cancer Res ; 41(1): 13, 2022 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-34996491

RESUMO

BACKGROUND: Increasing evidence has suggested inositol polyphosphate 5-phosphatase family contributes to tumorigenesis and tumor progression. However, the role of INPP5F in hepatocellular carcinoma (HCC) and its underlying mechanisms is unclear. METHODS: The expression of INPP5F in HCC was analyzed in public databases and our clinical specimens. The biological functions of INPP5F were investigated in vitro and vivo. The molecular mechanism of INPP5F in regulating tumor growth were studied by transcriptome-sequencing analysis, mass spectrometry analysis, immunoprecipitation assay and immunofluorescence assay. RESULTS: High expression of INPP5F was found in HCC tissues and was associated with poor prognosis in HCC patients. Overexpression of INPP5F promoted HCC cell proliferation, and vice versa. Knockdown of INPP5F suppressed tumor growth in vivo. Results from transcriptome-sequencing analysis showed INPP5F not only regulated a series of cell cycle related genes expression (c-MYC and cyclin E1), but also promoted many aerobic glycolysis related genes expression. Further studies confirmed that INPP5F could enhance lactate production and glucose consumption in HCC cell. Mechanistically, INPP5F activated Notch signaling pathway and upregulated c-MYC and cyclin E1 in HCC via interacting with ASPH. Interestingly, INPP5F was commonly nuclear-located in cells of adjacent non-tumor tissues, while in HCC, cytoplasm-located was more common. LMB (nuclear export inhibitor) treatment restricted INPP5F in nucleus and was associated with inhibition of Notch signaling and cell proliferation. Sequence of nuclear localization signals (NLSs) and nuclear export signals (NESs) in INPP5F aminoacidic sequence were then identified. Alteration of the NLSs or NESs influenced the localization of INPP5F and the expression of its downstream molecules. Furthermore, we found INPP5F interacted with both exportin and importin through NESs and NLSs, respectively, but the interaction with exportin was stronger, leading to cytoplasmic localization of INPP5F in HCC. CONCLUSION: These findings indicate that INPP5F functions as an oncogene in HCC via a translocation mechanism and activating ASPH-mediated Notch signaling pathway. INPP5F may serve as a potential therapeutic target for HCC patients.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Carcinoma Hepatocelular/genética , Inositol Polifosfato 5-Fosfatases/metabolismo , Neoplasias Hepáticas/genética , Proteínas de Membrana/metabolismo , Oxigenases de Função Mista/metabolismo , Proteínas Musculares/metabolismo , Animais , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Transdução de Sinais
12.
J Hepatocell Carcinoma ; 8: 985-996, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34466409

RESUMO

Ferroptosis is a special form of regulatory cell death caused by the accumulation of intracellular iron and lipid peroxidation. Here, we summarize the research progress on ferroptosis in hepatocellular carcinoma (HCC), trace the development of the concept of ferroptosis and its key regulatory factors, and discuss the application value of ferroptosis in the treatment of HCC from different perspectives. We believe that exploring the relationship between ferroptosis and HCC and clarifying the metabolism and expression of ferroptosis-specific genes and molecules will accelerate the development of novel ferroptosis-related molecules as HCC markers and therapeutic targets. We hope to provide a theoretical basis for better diagnosis and treatment to effectively improve the prognosis of patients with HCC.

13.
Blood ; 137(14): 1905-1919, 2021 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-33751108

RESUMO

Chromosome 13q deletion [del(13q)], harboring the miR-15a/16-1 cluster, is one of the most common genetic alterations in mature B-cell malignancies, which originate from germinal center (GC) and post-GC B cells. Moreover, miR-15a/16 expression is frequently reduced in lymphoma and multiple myeloma (MM) cells without del(13q), suggesting important tumor-suppressor activity. However, the role of miR-15a/16-1 in B-cell activation and initiation of mature B-cell neoplasms remains to be determined. We show that conditional deletion of the miR-15a/16-1 cluster in murine GC B cells induces moderate but widespread molecular and functional changes including an increased number of GC B cells, percentage of dark zone B cells, and maturation into plasma cells. With time, this leads to development of mature B-cell neoplasms resembling human extramedullary plasmacytoma (EP) as well as follicular and diffuse large B-cell lymphomas. The indolent nature and lack of bone marrow involvement of EP in our murine model resembles human primary EP rather than MM that has progressed to extramedullary disease. We corroborate human primary EP having low levels of miR-15a/16 expression, with del(13q) being the most common genetic loss. Additionally, we show that, although the mutational profile of human EP is similar to MM, there are some exceptions such as the low frequency of hyperdiploidy in EP, which could account for different disease presentation. Taken together, our studies highlight the significant role of the miR-15a/16-1 cluster in the regulation of the GC reaction and its fundamental context-dependent tumor-suppression function in plasma cell and B-cell malignancies.


Assuntos
Linfoma Difuso de Grandes Células B/genética , MicroRNAs/genética , Neoplasias de Plasmócitos/genética , Animais , Linfócitos B/metabolismo , Linfócitos B/patologia , Deleção Cromossômica , Transtornos Cromossômicos/genética , Transtornos Cromossômicos/patologia , Cromossomos Humanos Par 13/genética , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Humanos , Linfoma Difuso de Grandes Células B/patologia , Camundongos Endogâmicos C57BL , Família Multigênica , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Neoplasias de Plasmócitos/patologia , Plasmócitos/metabolismo , Plasmócitos/patologia , Plasmocitoma/genética , Plasmocitoma/patologia
14.
Oncogene ; 40(1): 28-45, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33051595

RESUMO

Hepatitis B x protein (HBx) affects cellular protein expression and participates in the tumorigenesis and progression of hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC). Metabolic reprogramming contributed to the HCC development, but its role in HBV-related HCC remains largely unclear. Tyrosine-protein phosphatase nonreceptor type 13 (PTPN13) is a significant regulator in tumor development, however, its specific role in hepatocarcinogenesis remains to be explored. Here, we found that decreased PTPN13 expression was associated with HBV/HBx. Patients with low PTPN13 expression showed a poor prognosis. Functional assays revealed that PTPN13 inhibited proliferation and tumorigenesis in vitro and in vivo. Further mechanistic studies indicated that HBx inhibited PTPN13 expression by upregulating the expression of DNMT3A and interacting with DNMT3A. Furthermore, we found that DNMT3A bound to the PTPN13 promoter (-343 to -313 bp) in an epigenetically controlled manner associated with elevated DNA methylation and then inhibited PTPN13 transcription. In addition, we identified IGF2BP1 as a novel PTPN13-interacting gene and demonstrated that PTPN13 influences c-Myc expression by directly and competitively binding to IGF2BP1 to decrease the intracellular concentration of functional IGF2BP1. Overexpressing PTPN13 promoted c-Myc mRNA degradation independent of the protein tyrosine phosphatase (PTP) activity of PTPN13. Importantly, we discovered that the PTPN13-IGF2BP1-c-Myc axis was important for cancer cell growth through promoting metabolic reprogramming. We verified the significant negative correlations between PTPN13 expression and c-Myc, PSPH, and SLC7A1 expression in clinical HCC tissue samples. In summary, our findings demonstrate that PTPN13 is a novel regulator of HBV-related hepatocarcinogenesis and may play an important role in HCC. PTPN13 may serve as a prognostic marker and therapeutic target in HBV-related HCC patients.


Assuntos
Carcinoma Hepatocelular/patologia , Hepatite B/complicações , Neoplasias Hepáticas/patologia , Proteína Tirosina Fosfatase não Receptora Tipo 13/genética , Proteínas de Ligação a RNA/genética , Transativadores/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Animais , Biomarcadores Tumorais/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/virologia , Proliferação de Células , Estudos de Coortes , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA Metiltransferase 3A , Progressão da Doença , Regulação para Baixo , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Hepatite B/genética , Hepatite B/metabolismo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/virologia , Camundongos , Transplante de Neoplasias , Prognóstico , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myc/genética , Estabilidade de RNA
15.
Cancer Res ; 81(3): 713-723, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33288657

RESUMO

Cisplatin chemotherapy is standard care for many cancers but is toxic to the kidneys. How this toxicity occurs is uncertain. In this study, we identified apurinic/apyrimidinic endonuclease 2 (APE2) as a critical molecule upregulated in the proximal tubule cells (PTC) following cisplatin-induced nuclear DNA and mitochondrial DNA damage in cisplatin-treated C57B6J mice. The APE2 transgenic mouse phenotype recapitulated the pathophysiological features of C-AKI (acute kidney injury, AKI) in the absence of cisplatin treatment. APE2 pulldown-MS analysis revealed that APE2 binds myosin heavy-Chain 9 (MYH9) protein in mitochondria after cisplatin treatment. Human MYH9-related disorder is caused by mutations in MYH9 that eventually lead to nephritis, macrothrombocytopenia, and deafness, a constellation of symptoms similar to the toxicity profile of cisplatin. Moreover, cisplatin-induced C-AKI was attenuated in APE2-knockout mice. Taken together, these findings suggest that cisplatin promotes AKI development by upregulating APE2, which leads to subsequent MYH9 dysfunction in PTC mitochondria due to an unrelated role of APE2 in DNA damage repair. This postulated mechanism and the availability of an engineered transgenic mouse model based on the mechanism of C-AKI provides an opportunity to identify novel targets for prophylactic treatment of this serious disease. SIGNIFICANCE: These results reveal and highlight an unexpected role of APE2 via its interaction with MYH9 and suggest that APE2 has the potential to prevent acute kidney injury in patients with cisplatin-treated cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/3/713/F1.large.jpg.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Antineoplásicos/efeitos adversos , Cisplatino/efeitos adversos , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Endonucleases/metabolismo , Túbulos Renais Proximais/efeitos dos fármacos , Enzimas Multifuncionais/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Injúria Renal Aguda/prevenção & controle , Animais , Carboplatina/efeitos adversos , Dano ao DNA , DNA Mitocondrial/efeitos dos fármacos , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/efeitos dos fármacos , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , Endonucleases/efeitos dos fármacos , Endonucleases/genética , Perda Auditiva Neurossensorial/induzido quimicamente , Humanos , Túbulos Renais Proximais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Doenças Mitocondriais/genética , Enzimas Multifuncionais/efeitos dos fármacos , Enzimas Multifuncionais/genética , Mutação , Cadeias Pesadas de Miosina/genética , Nefrite/induzido quimicamente , Oxaliplatina/efeitos adversos , Fenótipo , Trombocitopenia/induzido quimicamente , Regulação para Cima/efeitos dos fármacos
16.
Theranostics ; 10(22): 10345-10359, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32929353

RESUMO

Background: In addition to protein tyrosine kinases, accumulating evidence has shown that protein tyrosine phosphatases (PTPs) are suitable therapeutic targets in cancer. PRL-3 is a PTP member that has been well studied in many malignant tumours. The goal of the present study was to elucidate the role of PRL-3 in hepatocellular carcinoma (HCC), which remains largely unknown. Methods: Bioinformatic and immunohistochemical analyses were performed to analyse PRL-3 expression in HCC tissue samples and determine its clinical relevance. PRL-3 gene copy number variations were evaluated by bioinformatic analysis and quantitative-genomic polymerase chain reaction. The biological functions of PRL-3 were investigated in vivo and vitro. Gene microarray assays, RT-qPCR, western blotting and luciferase experiments were performed to identify the downstream effectors of PRL-3 that mediate its functions in HCC. Results: PRL-3 expression was upregulated in HCC samples from public databases and in cohort samples from our centre. High PRL-3 expression was associated with poor prognosis. Copy number gains and amplification of chromosome 8q24.3 in HCC were determined to be positively correlated with the PRL-3 overexpression. PRL-3 overexpression promoted HCC cell proliferation, migration and adhesion, while its loss had the opposite effects. Further study showed that focal adhesion kinase (FAK) was co-amplified and co-expressed with PRL-3 in HCC. Interestingly, PRL-3 also promoted the phosphorylation of FAK, which subsequently mediated the oncogenic functions of PRL-3 in HCC cells. Moreover, TGFB1 was identified as a downstream molecule of PRL-3. TGF-ß signalling was shown to mediate the PRL-3-induced activation of FAK. Furthermore, the p38 and PI3K/AKT pathways were observed to mediate the PRL-3-induced expression of TGFB1 and the subsequent activation of FAK, while the activation of FAK in turn stimulated activation of the p38 and PI3K/AKT pathways, forming a PRL-3-triggered AKT/p38/TGFB1/FAK positive feedback loop. Conclusion: Collectively, our findings indicate that the PTP PRL-3 plays a crucial role in the progression of HCC and provides an example of how co-amplified genes work together in HCC.


Assuntos
Carcinoma Hepatocelular/genética , Quinase 1 de Adesão Focal/genética , Neoplasias Hepáticas/genética , Proteínas de Neoplasias/genética , Proteínas Tirosina Fosfatases/genética , Animais , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Variações do Número de Cópias de DNA/genética , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Oncogenes/genética , Fosfatidilinositol 3-Quinases/genética , Fosforilação/genética , Prognóstico , Transdução de Sinais/genética , Regulação para Cima/genética
17.
Front Oncol ; 10: 1281, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32850391

RESUMO

Objective: Gallbladder cancer (GBC) is one of the most aggressive malignant tumors, and there is no effective and convenient method for predicting cancer-specific survival (CSS). We aim to develop a novel nomogram staging system based on the positive lymph node ratio (pLNR) for GBC patients. Methods:A total of 1,356 patients enrolled in the study. We evaluated the prognostic value of the pLNR and built a prognostic nomogram staging system based on the pLNR in the training cohort. The concordance index and calibration plots were used to evaluate model discrimination. The predictive accuracy and clinical value of the nomograms were measured by decision curve analysis (DCA). The CSS nomogram was further validated in an internal validation cohort. Results:The pLNR was an independent prognostic factor for CSS based on Cox regression analyses. A prognostic nomogram that combined T classification, pLNR, M classification, histologic grade, live metastasis, and tumor size was formulated with a c-index of 0.763 (95% CI, 0.728-0.798), while the c-indexes for the staging system of AJCC 8th, 7th, and 6th for CSS prediction were 0.718, 0.718, and 0.717, respectively. The calibration curves showed perfect agreement. The DCA showed that the nomogram provided substantial clinical value. The nomogram (the AUCs for 1, 3, and 5 years were 0.693, 0.716, and 0.726, respectively,) showed high prognostic accuracy. Conclusion:We have developed a formulated nomogram staging system based on the pLNR that allows more accurate individualized predictions of CSS for resected GBC patients than the AJCC staging systems.

18.
Cancer Cell Int ; 20: 140, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32368186

RESUMO

BACKGROUND: The primary tumor, regional lymph nodes and distant metastasis (TNM) stage is an independent risk factor for 1-year hepatocellular carcinoma (HCC) recurrence but has insufficient predictive efficiency. We attempt to develop and validate a nomogram to predict 1-year recurrence in HCC and improve the predictive efficiency of the TNM stage. METHODS: A total of 541 HCC patients were enrolled in the study. The risk score (RS) model was established with the logistic least absolute shrinkage and selector operation algorithm. The predictive nomogram was further validated in the internal testing cohort and external validation cohort. The area under the receiver operating characteristic curves (AUCs), decision curves and clinical impact curves were used to evaluate the predictive accuracy and clinical value of the nomogram. RESULTS: In the training cohort, we identified a RS model consisting of five stage-related genes (NUP62, EHMT2, RANBP1, MSH6 and FHL2) for recurrence at 1 year. The 1-year disease-free survival of patients was worse in the high-risk group than in the low-risk group (P < 0.0001), and 1-year recurrence was more likely in the high-risk group (Hazard ratio: 3.199, P < 0.001). The AUC of the nomogram was 0.739, 0.718 and 0.693 in the training, testing and external validation cohort, respectively, and these values were larger than the corresponding AUC of the TNM stage (0.681, 0.688 and 0.616, respectively). CONCLUSIONS: A RS model consisting of five stage-related genes was successfully identified for predicting 1-year HCC recurrence. Then, a novel nomogram based on the RS model and TNM stage to predict 1-year HCC recurrence was also developed and validated.

19.
Cancer Res ; 80(10): 2031-2044, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32193289

RESUMO

Multiple myeloma is an incurable refractory hematologic malignancy arising from plasma cells in the bone marrow. Here we investigated miR-26a function in multiple myeloma and tested single-wall carbon nanotube delivery of miR-26a in vitro and in vivo. miR-26a was downregulated in patients with multiple myeloma cells compared with plasma cells from healthy donors. miR-26a overexpression inhibited proliferation and migration and induced apoptosis in multiple myeloma cell lines. To identify the targets of miR-26a, RPMI8226-V-miR-26-GFP and RPMI8226-V-GFP cells were cultured using stable isotope labeling by amino acids in cell culture (SILAC) medium, followed by mass spectrometry analysis. In multiple myeloma cells overexpressing miR-26a, CD38 protein was downregulated and subsequently confirmed to be a direct target of miR-26a. Depletion of CD38 in multiple myeloma cells duplicated the multiple myeloma inhibition observed with exogenous expression of miR-26a, whereas restoration of CD38 overcame the inhibition of miR-26a in multiple myeloma cells. In a human multiple myeloma xenograft mouse model, overexpression of miR-26a inhibited CD38 expression, provoked cell apoptosis, and inhibited cell proliferation. Daratumumab is the first CD38 antibody drug for monotherapy and combination therapy for patients with multiple myeloma, but eventually resistance develops. In multiple myeloma cells, CD38 remained at low level during daratumumab treatment, but a high-quality response is sustained. In daratumumab-resistant multiple myeloma cells, CD38 expression was completely restored but failed to correlate with daratumumab-induced cell death. Therefore, a therapeutic strategy to confer selection pressure to maintain low CD38 expression in multiple myeloma cells may have clinical benefit. SIGNIFICANCE: These results highlight the tumor suppressor function of miR-26a via its targeting of CD38 and suggest the therapeutic potential of miR-26a in patients with multiple myeloma.


Assuntos
ADP-Ribosil Ciclase 1/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/genética , MicroRNAs/genética , Mieloma Múltiplo/patologia , ADP-Ribosil Ciclase 1/genética , Animais , Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Antineoplásicos Imunológicos/farmacologia , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Xenoenxertos , Humanos , Camundongos , MicroRNAs/farmacologia , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo
20.
Ann Surg Oncol ; 27(5): 1361-1371, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-31773517

RESUMO

BACKGROUND: The aim of the present work is to develop and validate accurate preoperative nomograms to predict microvascular invasion (MVI) and lymph node metastasis (LNM) in hepatocellular carcinoma. PATIENTS AND METHODS: A total of 268 patients with resected hepatocellular carcinoma (HCC) were divided into a training set (n = 180), in an earlier period, and a validation set (n = 88), thereafter. Risk factors for MVI and LNM were assessed based on logistic regression. Blood signatures were established using the least absolute shrinkage and selection operator algorithm. Nomograms were constructed by combining risk factors and blood signatures. Performance was evaluated using the training set and validated using the validation set. The clinical values of the nomograms were measured by decision curve analysis. RESULTS: The risk factors for MVI were hepatitis B virus (HBV) DNA loading, portal hypertension, Barcelona liver clinic (BCLC) stage, and three computerized tomography (CT) imaging features, namely tumor number, size, and encapsulation, while only BCLC stage, Child-Pugh classification, and tumor encapsulation were associated with LNM. The nomogram incorporating both risk factors and blood signatures achieved better performance in predicting MVI in the training and validation sets (C-indexes of 0.828 and 0.804) than the LNM nomogram (C-indexes of 0.765 and 0.717). Calibration curves also demonstrated a good fit. The decision curves indicate significant clinical usefulness. CONCLUSIONS: The novel validated nomograms for HCC patients presented herein are noninvasive preoperative tools that can effectively predict the individualized risk of MVI and LNM, and this predictive power can aid doctors in explaining the illness for patient counseling.


Assuntos
Vasos Sanguíneos/patologia , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Linfonodos/patologia , Nomogramas , Adulto , Fosfatase Alcalina/sangue , Aspartato Aminotransferases/sangue , Carcinoma Hepatocelular/sangue , Carcinoma Hepatocelular/complicações , Carcinoma Hepatocelular/diagnóstico por imagem , DNA Viral/sangue , Técnicas de Apoio para a Decisão , Feminino , Veias Hepáticas/patologia , Vírus da Hepatite B , Hepatite B Crônica/complicações , Humanos , Hipertensão Portal/complicações , Imageamento Tridimensional , Cirrose Hepática/complicações , Cirrose Hepática/diagnóstico por imagem , Neoplasias Hepáticas/sangue , Neoplasias Hepáticas/complicações , Neoplasias Hepáticas/diagnóstico por imagem , Modelos Logísticos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Veia Porta/patologia , Reprodutibilidade dos Testes , Estudos Retrospectivos , Fatores de Risco , Tomografia Computadorizada por Raios X , Carga Tumoral , Carga Viral , alfa-Fetoproteínas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA