Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
Front Immunol ; 13: 956224, 2022.
Article in English | MEDLINE | ID: mdl-36032075

ABSTRACT

Background: Cancer-associated fibroblasts (CAFs) are essential components of the tumor microenvironment (TME). These cells play a supportive role throughout cancer progression. Their ability to modulate the immune system has also been noted. However, there has been limited investigation of CAFs in the TME of epithelial ovarian cancer (EOC). Methods: We comprehensively evaluated the CAF landscape and its association with gene alterations, clinical features, prognostic value, and immune cell infiltration at the pan-cancer level using multi-omic data from The Cancer Genome Atlas (TCGA). The CAF contents were characterized by CAF scores based on the expression levels of seven CAF markers using the R package "GSVA." Next, we identified the molecular subtypes defined by CAF markers and constructed a CAF riskscore system using principal component analysis in the EOC cohort. The correlation between CAF riskscore and TME cell infiltration was investigated. The ability of the CAF riskscore to predict prognosis and immunotherapy response was also examined. Results: CAF components were involved in multiple immune-related processes, including transforming growth factor (TGF)-ß signaling, IL2-STAT signaling, inflammatory responses, and Interleukin (IL) 2-signal transducer and activator of transcription (STAT) signaling. Considering the positive correlation between CAF scores and macrophages, neutrophils, and mast cells, CAFs may exert immunosuppressive effects in both pan-cancer and ovarian cancer cohorts, which may explain accelerated tumor progression and poor outcomes. Notably, two distinct CAF molecular subtypes were defined in the EOC cohort. Low CAF riskscores were characterized by favorable overall survival (OS) and higher efficacy of immunotherapy. Furthermore, 24 key genes were identified in CAF subtypes. These genes were significantly upregulated in EOC and showed a strong correlation with CAF markers. Conclusions: Identifying CAF subtypes provides insights into EOC heterogeneity. The CAF riskscore system can predict prognosis and select patients who may benefit from immunotherapy. The mechanism of interactions between key genes, CAF markers, and associated cancer-promoting effects needs to be further elucidated.


Subject(s)
Cancer-Associated Fibroblasts , Ovarian Neoplasms , Carcinoma, Ovarian Epithelial , Female , Humans , Immunotherapy , Transforming Growth Factor beta , Tumor Microenvironment
3.
Cell Mol Gastroenterol Hepatol ; 12(2): 465-487, 2021.
Article in English | MEDLINE | ID: mdl-33667716

ABSTRACT

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) is the most common type of hepatic malignancies, with poor prognosis and low survival rate. Paraspeckles, which are unique subnuclear structures, are recently found to be involved in the development of various tumors, including HCC, and are related to induction in chemoresistance of HCC. This study aimed to investigate the possibility of paraspeckle in HCC cells participating in immune escape and its underlying mechanism in vitro and in vivo. METHODS: Expression of NEAT1_2, the framework of paraspeckle, in HCC cells and tissues was detected by qRT-PCR and RNA-FISH. mRNAs interacted with NEAT1_2 were pull-downed and sequenced in C-terminal S1-aptamer-tagged NEAT1_2 endogenously expressed HCC cells constructed using CRISPR-CAS9 knock-in technology. The effects of paraspeckle on HCC sensitivity to T-cell-mediated cytolysis were detected by T-cell mediated tumor cell killing assay. The roles of NEAT1_2 or NONO on IFNGR1 expression and IFN-γ signaling by applying gene function loss analysis in HCC cells were detected by qRT-PCR, RNA immunoprecipitation, Western blotting, and ELISA. The role of paraspeckle during adoptive T-cell transfer therapy for HCC in vivo was performed with a subcutaneous xenograft mouse. RESULTS: Paraspeckle in HCC cells is negatively related to T-cell-mediated cytolysis. Destruction of paraspeckle in HCC cells by knockdown of NEAT1_2 or NONO significantly improved the sensibility of resistant HCC cells to T-cell killing effects. Furthermore, IFNGR1 mRNA, which is sequestered by NEAT1_2 and NONO, is abundant in paraspeckle of T-cell killing-resistant HCC cells. Incapable IFN-γ-IFNGR1 signaling accounts for paraspeckle mediated-adoptive T-cell therapy resistance. Moreover, NEAT1_2 expression negatively correlates with IFNGR1 expression in clinical HCC tissues. CONCLUSIONS: Paraspeckle in HCC cells helps tumor cells escape from immunosurveillance through sequestering IFNGR1 mRNA to inhibiting IFN-γ-IFNGR1 signaling, thereby avoiding T-cell killing effects. Collectively, our results hint that NEAT1_2 highly expressed HCC patient is more resistant to T-cell therapy in clinic, and NEAT1_2 may be potential target for HCC immunotherapy.


Subject(s)
Carcinoma, Hepatocellular/immunology , Immune Evasion , Liver Neoplasms/immunology , Paraspeckles/metabolism , Receptors, Interferon/genetics , Adoptive Transfer , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Death , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Humans , Interferon-gamma/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/therapy , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Receptors, Interferon/metabolism , T-Lymphocytes/immunology , Interferon gamma Receptor
4.
Cancer Cell Int ; 20: 425, 2020.
Article in English | MEDLINE | ID: mdl-32884448

ABSTRACT

BACKGROUND: Dysregulation of fatty acid (FA) metabolism is involved in hepatocellular carcinoma (HCC) development. Non-POU domain-containing octamer binding protein (NONO), known as the component of nuclear paraspeckles, has recently been found to promote HCC progression. In this study, we investigated the functions of NONO in regulating de novo FA synthesis and its underling mechanism during HCC development. METHODS: The roles of NONO in HCC development by applying gene function loss analysis in HCC cells were detected by quantitative real-time polymerase chain reaction, cell proliferation, and cell invasion assays. The underlying mechanism of NONO in HCC development was examined by western blotting, subcellular fractionation, RNA-binding protein immunoprecipitation-sequencing, chromatin immunoprecipitation, co-immunoprecipitation and mass spectrometry. The effect of NONO on tumorigenesis in vivo was performed with a subcutaneous xenograft mouse model of HCC. RESULTS: NONO promotes HCC progression by interacting with and increasing ATP-citrate lyase (ACLY) mRNA to enhance FA biosynthesis. Furthermore, NONO promotes ACLY expression through enhancing nuclear ACLY mRNA stability in Diethylnitrosamine-stimulated HCC cells, not related to nuclear paraspeckles. Moreover, we find that NONO/SFPQ (Splicing factor proline and glutamine rich) heterodimer is essential for NONO interacting with ACLY mRNA in DEN stimulated HCC cells. In addition, NONO, Insulin like growth factor 2 mRNA binding protein 1 (IGF2BP1) and ACLY expressions contribute HCC development in mice and are related to poor survival. CONCLUSION: NONO promotes HCC progression by enhancing FA biosynthesis through interacting with ACLY mRNA and provide a novel potential target for HCC therapy.

5.
Pharmacol Res ; 157: 104825, 2020 07.
Article in English | MEDLINE | ID: mdl-32330553

ABSTRACT

Portal venous system thrombosis (PVST) is a life-threatening complication after splenectomy in cirrhotics patients with portal hypertension, while the application of intervention regimen may prevent the incidence of PVST. The aim of this network meta-analysis was to determine the most appropriate intervention regimen and application time. Several electronic databases were searched up to December 2019. We estimated summary odds ratios (OR) using pairwise and network meta-analyses with random effects for the outcome of occurrence of PVST. This work was registered with PROSPERO (CRD42019161406). The analysis was based on 19 researches, which included 1853 patients. The results drawn from the data in standard meta-analysis indicated that the application of intervention was better than no intervention use, and early application of interventions was better than delayed application in preventing the occurrence of PVST. Subsequent network meta-analysis was performed to determine the most suitable intervention regimen used early post-operation. For separate mono-therapy drug, alprostadil, antithrombin III, low molecular dextran were significantly more efficacious than others. However, mono-therapy analysis was not so close to clinical application. In the follow-up network meta-analysis, low molecular dextran combined with low molecular weight heparin exhibited the largest effect on the preventing the incidence of PVST (0.12, 0.03-0.49), followed by antithrombin III (0.12, 0.04-0.41) with low molecular dextran (0.14, 0.05-0.41). We could draw the conclusion that early application of low molecular weight heparin combined with low molecular dextran seems to be the most satisfactory treatment to prevent the incidence of PVST for patients with cirrhotic portal hypertension after splenectomy.


Subject(s)
Dextrans/therapeutic use , Fibrinolytic Agents/therapeutic use , Heparin, Low-Molecular-Weight/therapeutic use , Hypertension, Portal/surgery , Liver Cirrhosis/surgery , Portal Pressure , Portal Vein/physiopathology , Splenectomy/adverse effects , Venous Thrombosis/prevention & control , Dextrans/adverse effects , Drug Therapy, Combination , Fibrinolytic Agents/adverse effects , Heparin, Low-Molecular-Weight/adverse effects , Humans , Hypertension, Portal/diagnosis , Hypertension, Portal/physiopathology , Liver Cirrhosis/diagnosis , Liver Cirrhosis/physiopathology , Network Meta-Analysis , Protective Factors , Risk Assessment , Risk Factors , Treatment Outcome , Venous Thrombosis/diagnosis , Venous Thrombosis/etiology , Venous Thrombosis/physiopathology
6.
Cell Mol Biol Lett ; 25: 8, 2020.
Article in English | MEDLINE | ID: mdl-32123532

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is still the most common cause of cancer-related mortality worldwide and accumulating studies report that HCC is frequently linked to chronic inflammation. G-protein alpha-subunit (GNAS)-activating mutations have recently been reported to form a rare subgroup of inflammatory liver tumors. In this study, we investigated the roles of GNAS in inflammation-related HCC progression and its underlying mechanism. METHODS: Lipopolysaccharides (LPS) and diethylnitrosamine were employed to stimulate HCC cells to an induced inflammatory response. qRT-PCR, immunohistochemistry and immunoblotting were performed to detect the expression of GNAS in HCC tissues and cell lines. Expression levels of proinflammatory cytokines were detected by qRT-PCR and ELISA. N6-methyladenosine (m6A) methylation of GNAS mRNA was detected by RNA-binding protein immunoprecipitation (RIP). Transcription factors activation profiling plate array was performed to investigate the underlying mechanism in GNAS promoting interleukin-6 (IL-6) expression in HCC cells. HCC cell invasion was determined by transwell assay in vitro, and tumorigenesis was assessed with a subcutaneous xenograft mouse model of HCC. RESULTS: We found that LPS stimulation promotes GNAS expression in HCC cells through increasing m6A methylation of GNAS mRNA. The high expression level of GNAS promotes LPS-induced HCC cell growth and invasion by interacting with signal transducer and activator of transcription 3 (STAT3). Furthermore, GNAS knockdown inhibits LPS induced-IL-6 expression in HCC cells by suppressing STAT3 activation. Moreover, we found that GNAS promotes LPS-induced STAT3 activation in HCC cells through inhibiting long non-coding RNA TPTEP1 interacting with STAT3. In addition, GNAS expression promotes HCC development in mice and is related to poor survival. CONCLUSIONS: Our findings for the first time indicate a tumor-promoting role of GNAS in inflammation-related HCC progression and provide a novel potential target for HCC therapy.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Chromogranins/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Gene Expression Regulation, Neoplastic/genetics , Inflammation/metabolism , Liver Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Chromogranins/genetics , Disease Progression , GTP-Binding Protein alpha Subunits, Gs/genetics , Gene Knockdown Techniques , Humans , Inflammation/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Methylation , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , RNA Interference , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Signal Transduction/genetics , Xenograft Model Antitumor Assays
7.
J Exp Clin Cancer Res ; 38(1): 189, 2019 May 09.
Article in English | MEDLINE | ID: mdl-31072375

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is still the most common cause of tumor-related death worldwide and accumulating studies report that long non-coding RNAs (LncRNAs) are closely related with HCC development, metastasis and prognosis. Cisplatinum, a well-known chemotherapeutic drug, has been widely used for treatment of numerous human cancers including HCC. This study aimed to investigate the differential expressions of LncRNAs in HCC cells treated with cisplatinum and its underlying mechanism. METHODS: The differential expressions of LncRNAs in HCC cells treated with cisplatinum were determined by RNA-seq. The roles of TPTEP1 in HCC development by applying gene function gain and loss analysis in MHCC97H and QYG-7703 cell lines were detected by quantitative real-time polymerase chain reaction (qRT-PCR), cell proliferation, colony formation, cell invasion and flow cytometry assays. The underlying mechanism of TPTEP1 sensitizing hepatocellular carcinoma cells to cisplatinum was examined by RNA-pull down, western blotting, subcellular fractionation, RNA immunoprecipitation and dual luciferase reporter assays. The effect of TPTEP1 on tumorigenesis in vivo was performed with a subcutaneous xenograft mouse model of HCC. In addition, TPTEP1 expression was detected in clinical tumor tissue samples by qRT-PCR. RESULTS: LncRNA TPTEP1 was highly expressed in cisplatinum-treated HCC cells, which sensitizes hepatocellular carcinoma cell to cisplatinum-induced apoptosis. TPTEP1 overexpression inhibited, while TPTEP1 knockdown promoted HCC cell proliferation, tumorigenicity and invasion. Furthermore, TPTEP1 exerted its tumor suppressing activities by interacting with signal transducer and activator of transcription 3 (STAT3) to inhibit its phosphorylation, homodimerization, nuclear translocation and down-stream genes transcription. Moreover, TPTEP1 overexpression obviously inhibits tumor masses in vivo in a subcutaneous xenograft mouse model of HCC and TPTEP1 is frequently downregulated in HCC tissues, compared to its corresponding pre-tumor tissues. CONCLUSION: LncRNA TPTEP1 inhibits hepatocellular carcinoma cells progression by affecting IL-6/STAT3 signaling. Taken together, our findings suggest a tumor suppressing role of TPTEP1 in HCC progression and provide a novel understanding of TPTEP1 during the chemotherapy for HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Cisplatin/administration & dosage , Liver Neoplasms/drug therapy , RNA, Long Noncoding/genetics , STAT3 Transcription Factor/genetics , Animals , Apoptosis/drug effects , Carcinogenesis/drug effects , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-6/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Phosphorylation , Prognosis , Xenograft Model Antitumor Assays
8.
Biochem Biophys Res Commun ; 500(4): 973-980, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29715456

ABSTRACT

Long noncoding RNA (LncRNA) PVT1 has recently been reported to be involved in the development of hepatocellular carcinoma (HCC) and hsigh expression of oncogenic PVT1 is associated with poor prognosis of HCC. Interferon-α (IFN-α) has been used in clinic for HCC therapy. However, whether PVT1 is involved in the IFN-α therapy for HCC is completely unknown. Our study found that high PVT1 expression in HCC cells is associated with high unmethylation in PVT1 promoter region. IFN-α treatment further increases PVT1 expression in HCC cells by enhancing H3K4me3 modification on the promoter. Furthermore, PVT1 knockdown enhances IFN-α-induced HCC cell apoptosis by promoting phosphorylation of signal transducer and activator of transcription 1 (STAT1) and upregulating IFN-stimulated genes expression. Moreover, PVT1 specifically interacts with STAT1 in HCC cells. Taken together, these results for the first time indicate that IFN-α treatment promotes oncogenic PVT1 expression in HCC cells, which interacts with STAT1 to inhibit IFN-α signaling, ultimately blocking IFN-α-induced cells apoptosis, suggesting that lncRNA PVT1 may be a potential target to improve IFN-α-mediated HCC immunotherapies.


Subject(s)
Gene Expression Regulation, Neoplastic , Histones/genetics , Interferon-alpha/pharmacology , RNA, Long Noncoding/genetics , STAT1 Transcription Factor/genetics , Apoptosis/drug effects , Cell Line, Tumor , Hep G2 Cells , Histones/metabolism , Humans , Interferon alpha-2 , Phosphorylation , Promoter Regions, Genetic , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Proteins/pharmacology , STAT1 Transcription Factor/metabolism , Signal Transduction
9.
Chem Pharm Bull (Tokyo) ; 66(5): 535-540, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29515052

ABSTRACT

Twelve pseudo-ginsenosides were synthesized under a mild condition, via a simple three-step called acetylation, elimination-addition and saponification. The inhibitory effects of these twelve pseudo-ginsenosides were screened on the hemolysis of rabbit erythrocytes caused by 2,2'-azobis (2-amidinopropane hydrochloride) (AAPH). It was found that the IC50 values followed the sequence of (20Z) pseudo-protopanaxatriol (pseudo-PPT)<(20Z) pseudo-protopanaxadiol (pseudo-PPD)<(20Z) pseudo-Rh2<(20E) pseudo-PPT<(20E) pseudo-PPD<(20E) pseudo-Rh2<(20Z) pseudo-Rg2<(20E) pseudo-Rg2

Subject(s)
Antioxidants/chemical synthesis , Antioxidants/pharmacology , Erythrocytes/drug effects , Ginsenosides/chemical synthesis , Ginsenosides/pharmacology , Amidines/antagonists & inhibitors , Amidines/pharmacology , Animals , Antioxidants/chemistry , Dose-Response Relationship, Drug , Free Radicals/chemistry , Ginsenosides/chemistry , Hemolysis/drug effects , Molecular Conformation , Rabbits , Stereoisomerism , Structure-Activity Relationship
10.
Nat Prod Commun ; 11(5): 601-3, 2016 May.
Article in English | MEDLINE | ID: mdl-27319127

ABSTRACT

Two ginsenoside derivatives (1, 2) along with 2 known ginsenosides (3, 4) were isolated from the acid hydrolysis products of pseudoginsenoside-F11. Their structures were elucidated on the basis of spectroscopic analyses, including ID, 2D NMR and HR-ESI-MS. Among them, (12R, 20S, 24S)-20, 24; 12, 24-diepoxy-dammarane-3ß, 6α-diol (1) and (20R, 24R)-dammar-20, 24-epoxy-3ß, 6α, 12ß, 25-tetraol (2) were identified as new triterpenoid saponins. They were subjected to assay for cytotoxic activities against six human tumor cells lines.


Subject(s)
Ginsenosides/chemistry , Saponins/chemical synthesis , Triterpenes/chemical synthesis , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...