Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
2.
Adv Sci (Weinh) ; 10(2): e2202448, 2023 01.
Article in English | MEDLINE | ID: mdl-36453576

ABSTRACT

The guanine nucleotide exchange factor (GEF) SOS1 catalyzes the exchange of GDP for GTP on RAS. However, regulation of the GEF activity remains elusive. Here, the authors report that PPDPF functions as an important regulator of SOS1. The expression of PPDPF is significantly increased in pancreatic ductal adenocarcinoma (PDAC), associated with poor prognosis and recurrence of PDAC patients. Overexpression of PPDPF promotes PDAC cell growth in vitro and in vivo, while PPDPF knockout exerts opposite effects. Pancreatic-specific deletion of PPDPF profoundly inhibits tumor development in KRASG12D -driven genetic mouse models of PDAC. PPDPF can bind GTP and transfer GTP to SOS1. Mutations of the GTP-binding sites severely impair the tumor-promoting effect of PPDPF. Consistently, mutations of the critical amino acids mediating SOS1-PPDPF interaction significantly impair the GEF activity of SOS1. Therefore, this study demonstrates a novel model of KRAS activation via PPDPF-SOS1 axis, and provides a promising therapeutic target for PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Mice , Carcinoma, Pancreatic Ductal/genetics , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Guanosine Triphosphate , Intracellular Signaling Peptides and Proteins , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , SOS1 Protein , Pancreatic Neoplasms
3.
Oncogene ; 41(36): 4244-4256, 2022 09.
Article in English | MEDLINE | ID: mdl-35906391

ABSTRACT

Lung cancer is the most common malignancy and the leading cause of cancer death worldwide, and lung adenocarcinoma (LUAD) is the most prevalent subtype. Considering the emergence of resistance to therapies, it is urgent to develop more effective therapies to improve the prognosis. Here we reported that pancreatic progenitor cell differentiation and proliferation factor (PPDPF) deficiency inhibited LUAD development both in vitro and in vivo. Mechanistically, PPDPF induces hyperactive STAT3 by interfering STAT3-PTPN1 interaction. Activated STAT3 promoted BMPR2 transcription, which further inhibited apoptosis. Moreover, PPDPF reduced NK cell infiltration and activation to develop an immunosuppressive microenvironment, which was also mediated by STAT3. Furthermore, we identified that the expression of PPDPF was positively correlated with the malignant features of LUAD, as well as BMPR2 and p-STAT3 level in clinical samples. Therefore, our study suggests that PPDPF positively regulates BMPR2 expression and facilitates immune escape via regulating STAT3 activity, providing a potential therapy target for LUAD.


Subject(s)
Adenocarcinoma of Lung , Adenocarcinoma , Lung Neoplasms , Adenocarcinoma/pathology , Adenocarcinoma of Lung/pathology , Apoptosis/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins , Killer Cells, Natural/metabolism , Lung Neoplasms/pathology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Tumor Microenvironment
4.
Cell Mol Gastroenterol Hepatol ; 14(1): 101-127, 2022.
Article in English | MEDLINE | ID: mdl-35390516

ABSTRACT

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) is a highly heterogeneous solid tumor with high morbidity and mortality. AT-rich interaction domain 1A (ARID1A) accounts for up to 10% of mutations in liver cancer, however, its role in HCC remains controversial, and no targeted therapy has been established. METHODS: The expression of ARID1A in clinical samples was examined by Western blot and immunohistochemical staining. ARID1A was knocked out by Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) in HCC cell lines, and the effects of glucose deprivation on cell viability, proliferation, and apoptosis were measured. Mass spectrometry analysis was used to find ARID1A-interacting proteins, and the result was verified by co-immunoprecipitation and Glutathione S Transferase (GST) pull-down. The regulation of ARID1A target gene USP9X was investigated by chromatin immunoprecipitation, Glutathione S Transferase (GST) pull-down, luciferase reporter assay, and so forth. Finally, drug treatments were performed to explore the therapeutic potential of the agents targeting ARID1A-deficient HCC in vitro and in vivo. RESULTS: Our study has shown that ARID1A loss protected cells from glucose deprivation-induced cell death. A mechanism study disclosed that AIRD1A recruited histone deacetylase 1 via its C-terminal region DUF3518 to the promoter of USP9X, resulting in down-regulation of USP9X and its target protein kinase AMP-activated catalytic subunit α2 (PRKAA2). ARID1A knockout and a 1989∗ truncation mutant in HCC abolished this effect, increased the levels of H3K9 and H3K27 acetylation at the USP9X promoter, and up-regulated the expression of USP9X and protein kinase AMP-activated catalytic subunit α2 (PRKAA2), which mediated the adaptation of tumor cells to glucose starvation. Compound C dramatically inhibited the growth of ARID1A-deficient tumors and prolongs the survival of tumor-bearing mice. CONCLUSIONS: HCC patients with ARID1A mutation may benefit from synthetic lethal therapy targeting the ubiquitin-specific peptidase 9 X-linked (USP9X)-adenosine 5'-monophosphate-activated protein kinase (AMPK) axis.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , AMP-Activated Protein Kinases , Adenosine Monophosphate , Animals , Carcinoma, Hepatocellular/pathology , Cell Proliferation/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Glucose , Glutathione Transferase , Humans , Liver Neoplasms/pathology , Mice , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin Thiolesterase/genetics
5.
Oncogene ; 40(34): 5302-5313, 2021 08.
Article in English | MEDLINE | ID: mdl-34247189

ABSTRACT

The prognosis of hepatocellular carcinoma (HCC) remains unsatisfactory due to limited effective treatment options. In this work, we investigated the therapeutic efficacy of Terbinafine for HCC and the underlying mechanism. The influence of Terbinafine on cell growth, 3D spheroid formation, clonogenic survival, and protein synthesis was investigated in human HCC cell lines. Co-immunoprecipitation, immunofluorescence, and other techniques were employed to explore how Terbinafine exerts its anticancer effect. Subcutaneous tumorigenicity assay, orthotopic and patient-derived xenograft (PDX) HCC models were used to evaluate the anticancer effect of Terbinafine monotherapy and the combinatorial treatment with Terbinafine and sorafenib against HCC. The anticancer activity of Terbinafine was Squalene epoxidase (SQLE)-independent. Instead, Terbinafine robustly suppressed the proliferation of HCC cells by inhibiting mTORC1 signaling via activation of AMPK. Terbinafine alone or in combination with sorafenib delayed tumor progression and markedly prolonged the survival of tumor-bearing mice. The synergy between Terbinafine and sorafenib was due to concomitant inhibition of mTORC1 and induction of severe persistent DNA double-strand breaks (DSBs), which led to the delayed proliferation and accelerated cell death. Terbinafine showed promising anticancer efficacy in preclinical models of HCC and may serve as a potential therapeutic strategy for HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , AMP-Activated Protein Kinases , Animals , Mice , Terbinafine
6.
Nat Commun ; 12(1): 3059, 2021 05 24.
Article in English | MEDLINE | ID: mdl-34031390

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) has become the most prevalent chronic liver disease in the world, however, no drug treatment has been approved for this disease. Thus, it is urgent to find effective therapeutic targets for clinical intervention. In this study, we find that liver-specific knockout of PPDPF (PPDPF-LKO) leads to spontaneous fatty liver formation in a mouse model at 32 weeks of age on chow diets, which is enhanced by HFD. Mechanistic study reveals that PPDPF negatively regulates mTORC1-S6K-SREBP1 signaling. PPDPF interferes with the interaction between Raptor and CUL4B-DDB1, an E3 ligase complex, which prevents ubiquitination and activation of Raptor. Accordingly, liver-specific PPDPF overexpression effectively inhibits HFD-induced mTOR signaling activation and hepatic steatosis in mice. These results suggest that PPDPF is a regulator of mTORC1 signaling in lipid metabolism, and may be a potential therapeutic candidate for NAFLD.


Subject(s)
Fatty Liver/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Cullin Proteins/metabolism , DNA-Binding Proteins/metabolism , HEK293 Cells , Hep G2 Cells , Humans , Lipid Metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/metabolism
7.
Proc Natl Acad Sci U S A ; 117(9): 4770-4780, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32071245

ABSTRACT

Recurrence and metastasis remain the major obstacles to successful treatment of hepatocellular carcinoma (HCC). Chromatin remodeling factor ARID2 is commonly mutated in HCC, indicating its important role in cancer development. However, its role in HCC metastasis is largely elusive. In this study, we find that ARID2 expression is significantly decreased in metastatic HCC tissues, showing negative correlation with pathological grade, organ metastasis and positive association with survival of HCC patients. ARID2 inhibits migration and invasion of HCC cells in vitro and metastasis in vivo. Moreover, ARID2 knockout promotes pulmonary metastasis in different HCC mouse models. Mechanistic study reveals that ARID2 represses epithelial-mesenchymal transition (EMT) of HCC cells by recruiting DNMT1 to Snail promoter, which increases promoter methylation and inhibits Snail transcription. In addition, we discover that ARID2 mutants with disrupted C2H2 domain lose the metastasis suppressor function, exhibiting a positive association with HCC metastasis and poor prognosis. In conclusion, our study reveals the metastasis suppressor role as well as the underlying mechanism of ARID2 in HCC and provides a potential therapeutic target for ARID2-deficient HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Chromatin Assembly and Disassembly/physiology , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Liver Neoplasms/drug therapy , Neoplasm Metastasis/drug therapy , Transcription Factors/metabolism , Animals , CYS2-HIS2 Zinc Fingers , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Disease Models, Animal , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Knockout , Mutation , Neoplasm Metastasis/pathology , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
8.
Oncogene ; 38(48): 7281-7293, 2019 11.
Article in English | MEDLINE | ID: mdl-31417183

ABSTRACT

Bone morphogenetic protein 10 (BMP10), one member of the BMP family, is involved in various development events. Dysregulation of BMP10 has been observed in several diseases, including hypertensive cardiac hypertrophy, Hirschsprung disease and blood vessel formation. However, its role in liver cancer remains largely unknown. In this study, we reported that BMP10 was significantly downregulated in HCC at both mRNA and protein level. Decreased BMP10 was associated with bigger tumor size, worse TNM stage, earlier recurrence and poorer survival. BMP10 negatively regulated HCC cell proliferation in vitro and in vivo. Mechanism study revealed that BMP10 suppressed tumor cell growth by inhibiting STAT3 signaling. Interestingly, we found that cytoplasmic BMP10 interacted with both receptor protein tyrosine phosphatase sigma (PTPRS) and STAT3, which facilitated dephosphorylation of STAT3 by PTPRS. Altogether, our study has revealed the clinical significance of BMP10 in HCC, and suppression of HCC cell growth by BMP10 via PTPRS-STAT3 axis, providing a potential therapeutic strategy for targeting STAT3 signaling in HCC.


Subject(s)
Biomarkers, Tumor/metabolism , Bone Morphogenetic Proteins/metabolism , Carcinoma, Hepatocellular/pathology , Gene Expression Regulation, Neoplastic , Liver Neoplasms/pathology , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , STAT3 Transcription Factor/metabolism , Adult , Animals , Apoptosis , Biomarkers, Tumor/genetics , Bone Morphogenetic Proteins/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Disease Progression , Female , Follow-Up Studies , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Mice, Nude , Neoplasm Metastasis , Phosphorylation , Prognosis , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , STAT3 Transcription Factor/genetics , Signal Transduction , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Nat Commun ; 10(1): 2510, 2019 06 07.
Article in English | MEDLINE | ID: mdl-31175290

ABSTRACT

Metastasis-associated recurrence is the major cause of poor prognosis in hepatocellular carcinoma (HCC), however, the underlying mechanisms remain largely elusive. In this study, we report that expression of choroideremia-like (CHML) is increased in HCC, associated with poor survival, early recurrence and more satellite nodules in HCC patients. CHML promotes migration, invasion and metastasis of HCC cells, in a Rab14-dependent manner. Mechanism study reveals that CHML facilitates constant recycling of Rab14 by escorting Rab14 to the membrane. Furthermore, we identify several metastasis regulators as cargoes carried by Rab14-positive vesicles, including Mucin13 and CD44, which may contribute to metastasis-promoting effects of CHML. Altogether, our data establish CHML as a potential promoter of HCC metastasis, and the CHML-Rab14 axis may be a promising therapeutic target for HCC.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Neoplasms, Multiple Primary/metabolism , rab GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/secondary , HEK293 Cells , Humans , Hyaluronan Receptors/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Nude , Mucins/metabolism , Neoplasm Invasiveness , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Transplantation , Neoplasms, Multiple Primary/pathology , RNA, Messenger/metabolism , Tumor Burden
10.
Cell Prolif ; 52(3): e12583, 2019 May.
Article in English | MEDLINE | ID: mdl-30793395

ABSTRACT

OBJECTIVES: Wnt1-inducible signalling pathway protein 3 (WISP3/CCN6) belongs to the CCN (CYR61/CTGF/NOV) family of proteins, dysregulation of this family contributed to the tumorigenicity of various tumours. In this study, we need to explore its role in hepatocellular carcinoma that remains largely elusive. MATERIALS AND METHODS: The expression of WISP3/CCN6 was analysed by qRT-PCR and Western blotting. Effects of WISP3 on proliferation and metastasis of HCC cells were examined, respectively, by MTT assay and Boyden Chamber. Roles of WISP3 on HCC tumour growth and metastatic ability in vivo were detected in nude mice. Related mechanism study was confirmed by immunofluorescence and Western blotting. RESULTS: The expression of WISP3 was significantly downregulated in HCC clinical samples and cell lines, and reversely correlated with the tumour size. Forced expression of WISP3 in HCC cells significantly suppressed cell growth and migration in vitro as well as tumour growth and metastatic seeding in vivo. In contrast, downregulation of WISP3 accelerated cell proliferation and migration, and promoted in vivo metastasis. Further study revealed that WISP3 inhibited the translocation of ß-catenin to the nucleus by activating glycogen synthase kinase-3ß (GSK3ß). Moreover, constitutively active ß-catenin blocked the suppressive effects of WISP3 on HCC. CONCLUSIONS: Our study showed that WISP3 suppressed the progression of HCC by negative regulation of ß-catenin/TCF/LEF signalling, providing WISP3 as a potential therapeutic candidate for HCC.


Subject(s)
CCN Intercellular Signaling Proteins/genetics , CCN Intercellular Signaling Proteins/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Active Transport, Cell Nucleus , Animals , CCN Intercellular Signaling Proteins/antagonists & inhibitors , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Down-Regulation , Gene Knockdown Techniques , Glycogen Synthase Kinase 3 beta/metabolism , Heterografts , Humans , Male , Mice , Mice, Nude , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Signal Transduction , TCF Transcription Factors/metabolism , beta Catenin/metabolism
11.
Cell Physiol Biochem ; 47(4): 1643-1654, 2018.
Article in English | MEDLINE | ID: mdl-29945155

ABSTRACT

BACKGROUND/AIMS: Although it has been widely accepted that protein arginine methyltransferase 1 (PRMT1) is a cancer-promoting gene in various cancers, the mechanism of PRMT1 in hepatocellular carcinoma (HCC) requires more exploration. This study aimed to investigate the role of PRMT1 in HCC growth and metastasis. METHODS: We compared PRMT1 expression and clinicopathological characteristics using paired HCC and adjacent noncancerous liver tissues from 210 patients and immunohistochemistry analyses. Cell proliferation, colony formation and migration were determined in HCC cell lines with PRMT1 overexpression or downregulation through MTT, crystal violet and Boyden chamber assays. Tumour growth was monitored in a xenograft model, and intrahepatic metastasis models were established. RESULTS: PRMT1 expression was greatly increased in clinical HCC samples and strongly associated with poor prognosis and recurrence; PRMT1 expression was also positively correlated with microvascular invasion (P = 0.024), tumour differentiation (P = 0.014), tumour size (P = 0.002), and portal vein tumour thrombus (PVTT) (P = 0.028). Cell proliferation, colony formation and migration in vitro were enhanced by PRMT1 upregulation and decreased by PRMT1 downregulation in HCC cell lines. Moreover, low PRMT1 expression resulted in slow tumour growth and decreased tumour weight in vivo, as well as tumour metastasis. These phenotypes were associated with STAT3 signalling pathway activation. Cryptotanshinone, a STAT3 inhibitor, inhibited STAT3 phosphorylation and reversed the HCC phenotype of PRMT1 expression. CONCLUSIONS: We revealed a significant role for PRMT1 in HCC progression and metastasis in vitro and in vivo via STAT3 signalling pathway activation. PRMT1 may be a potential novel prognostic biomarker and new therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Neoplasm Proteins/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Carcinoma, Hepatocellular/pathology , Female , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Male , Neoplasm Metastasis
12.
Cancer Med ; 7(6): 2629-2640, 2018 06.
Article in English | MEDLINE | ID: mdl-29733540

ABSTRACT

Adipocyte fatty acid-binding protein (FABP4) is abundant in macrophage and adipocyte. It is known to be involved in lipid metabolism. The role of FABP4 has been reported in various cancers, such as non-small cell lung cancer, breast cancer, ovarian cancer, and prostatic cancer. However, its role remains unclear in hepatocellular carcinoma (HCC). In our study, we investigated the expression of FABP4 at both mRNA and protein levels, and by examining 175 cases of patients with cancer of the liver tissue microarray, the significance between the expression of FABP4 and clinical characteristics had been discussed. We found that FABP4 was lowly expressed in HCC tissues compared to the corresponding tissue adjacent, and the expression of FABP4 was significantly associated with the tumor size, PVTT, recurrence-free survival and overall survival. Moreover, multivariate Cox regression analysis indicated that the expression of FABP4, Alb, AFP, HBsAg, and PVTT were independent risk factors for overall survival, and the expression of FABP4, AFP, GGT, tumor size, and encapsulation were independent risk factors for HCC recurrence. In addition, we revealed that FABP4 suppressed HCC cell proliferation and invasion in vitro. Moreover, overexpression of FABP4 led to inhibit tumor growth and decreased tumor volume in vivo. These phenotypes were associated with altered expression of Snail and p-STAT3. Our studies thus suggest that FABP4 could be a potential target for HCC chemotherapy.


Subject(s)
Biomarkers, Tumor , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Fatty Acid-Binding Proteins/genetics , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Adult , Aged , Animals , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/therapy , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Models, Animal , Fatty Acid-Binding Proteins/metabolism , Female , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Liver Neoplasms/diagnosis , Liver Neoplasms/therapy , Male , Mice , Middle Aged , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism
13.
J Exp Med ; 215(1): 177-195, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29237705

ABSTRACT

Although cilia loss and cell transformation are frequently observed in the early stage of tumorigenesis, the roles of cilia in cell transformation are unknown. In this study, disrupted ciliogenesis was observed in cancer cells and pancreatic cancer tissues, which facilitated oncogene-induced transformation of normal pancreatic cells (HPDE6C7) and NIH3T3 cells through activating the mevalonate (MVA) pathway. Disruption of ciliogenesis up-regulated MVA enzymes through ß catenin-T cell factor (TCF) signaling, which synchronized with sterol regulatory element binding transcription factor 2 (SREBP2), and the regulation of MVA by ß-catenin-TCF signaling was recapitulated in a mouse model of pancreatic ductal adenocarcinoma (PDAC) and human PDAC samples. Moreover, disruption of ciliogenesis by depleting Tg737 dramatically promoted tumorigenesis in the PDAC mouse model, driven by KrasG12D , which was inhibited by statin, an inhibitor of the MVA pathway. Collectively, this study emphasizes the crucial roles of cilia in governing the early steps of the transformation by activating the MVA pathway, suggesting that statin has therapeutic potential for pancreatic cancer treatment.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cilia/pathology , Metabolic Networks and Pathways , Mevalonic Acid/metabolism , Animals , Carcinoma, Pancreatic Ductal/etiology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Gene Expression Profiling , Humans , Male , Mice , NIH 3T3 Cells , Oncogenes , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Sterol Regulatory Element Binding Protein 2 , TCF Transcription Factors/metabolism , Pancreatic Neoplasms
14.
Biochem Biophys Res Commun ; 482(4): 1048-1053, 2017 Jan 22.
Article in English | MEDLINE | ID: mdl-27908734

ABSTRACT

Metabolic dysregulation is one of the most common and recognizable features of cancer. Triosephosphate isomerase 1 (TPI1), which catalyzes the interconversion of dihydroxyacetone phosphate (DHAP) and d-glyceraldehyde-3-phosphate (G3P) during glycosis and gluconeogenesis, is a crucial enzyme in the carbohydrate metabolism. However, the biological function and mechanism of TPI1 in cancer remain largely unknown. In this study, we have found that TPI1 expression was greatly decreased in clinical HCC samples, positively correlated with overall survival, and negatively associated with histological differentiation, tumor size and organ metastasis. Forced expression of TPI1 in HCC cells inhibited cell growth, migration, and invasion in vitro. Consistently, knockdown of TPI1 by shRNA promoted cell growth, migration and invasion. Moreover, overexpression of TPI1 led to slowed tumor growth and decreased tumor weight in vivo. Furthermore, cell cycle arrest was induced by TPI1 overexpression. These phenotypes were associated with altered expression of ß-catenin, Vimentin, P53, P27 and CyclinD1. Therefore, our data suggested that TPI1 functioned as a tumor suppressor in HCC and might serve as a potential therapeutic target for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , Triose-Phosphate Isomerase/metabolism , Aged , Animals , Carcinogenesis , Cell Movement , Cell Proliferation , Cyclin D1/metabolism , Disease Progression , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Transplantation , Proliferating Cell Nuclear Antigen/metabolism , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/metabolism , Vimentin/metabolism , beta Catenin/metabolism
15.
Hepatology ; 62(6): 1791-803, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26257239

ABSTRACT

UNLABELLED: Sorafenib is a specific adenosine triphosphate-competitive RAF inhibitor used as a first-line treatment of advanced hepatocellular carcinoma (HCC). However, the responses are variable, reflecting heterogeneity of the disease, while the resistance mechanism remains poorly understood. Here, we report that sorafenib treatment can exacerbate disease progression in both patient-derived xenografts and cell line-derived xenografts and that the therapeutic effect of the drug inversely covaries to the ratio of epithelial cell adhesion molecule-positive cells, which may be tumor initiating cells in HCC. The TSC2-AKT cascade mediates this sorafenib resistance. In response to sorafenib treatment, formation of the TSC1/2 complex is enhanced, causing increased phosphorylation of AKT, which contributes to up-regulation of "stemness"-related genes in epithelial cell adhesion molecule-positive cells and enhancement of tumorigenicity. The expression of TSC2 negatively correlated with prognosis in clinical sorafenib therapy. Furthermore, all-trans retinoic acid decreased AKT activity, reduced the epithelial cell adhesion molecule-positive cell population enriched by sorafenib, and potentiated the therapeutic effect of sorafenib in the patient-derived xenograft model. CONCLUSION: Our findings suggest that a subtype of HCC is not suitable for sorafenib therapy; this resistance to sorafenib can be predicted by the status of TSC2, and agents inducing differentiation of tumor initiating cells (e.g., all-trans retinoic acid) should improve the prognosis of this subtype of HCC.


Subject(s)
Antigens, Neoplasm/drug effects , Antineoplastic Agents/adverse effects , Carcinoma, Hepatocellular/chemically induced , Cell Adhesion Molecules/drug effects , Liver Neoplasms/chemically induced , Neoplastic Stem Cells/drug effects , Niacinamide/analogs & derivatives , Oncogene Protein v-akt/physiology , Phenylurea Compounds/adverse effects , Tumor Suppressor Proteins/physiology , Animals , Carcinoma, Hepatocellular/classification , Disease Progression , Epithelial Cell Adhesion Molecule , Humans , Liver Neoplasms/classification , Mice , Niacinamide/adverse effects , Sorafenib , Tuberous Sclerosis Complex 2 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...