Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 161(3): 595-609, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25892225

ABSTRACT

Organisms must be able to respond to low oxygen in a number of homeostatic and pathological contexts. Regulation of hypoxic responses via the hypoxia-inducible factor (HIF) is well established, but evidence indicates that other, HIF-independent mechanisms are also involved. Here, we report a hypoxic response that depends on the accumulation of lactate, a metabolite whose production increases in hypoxic conditions. We find that the NDRG3 protein is degraded in a PHD2/VHL-dependent manner in normoxia but is protected from destruction by binding to lactate that accumulates under hypoxia. The stabilized NDRG3 protein binds c-Raf to mediate hypoxia-induced activation of Raf-ERK pathway, promoting angiogenesis and cell growth. Inhibiting cellular lactate production abolishes the NDRG3-mediated hypoxia responses. Our study, therefore, elucidates the molecular basis for lactate-induced hypoxia signaling, which can be exploited for the development of therapies targeting hypoxia-induced diseases.


Subject(s)
Hypoxia/metabolism , Lactic Acid/metabolism , Cell Hypoxia , Cell Line , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Intracellular Signaling Peptides and Proteins , MAP Kinase Signaling System , Neovascularization, Pathologic/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Oxygen/metabolism , Protein Binding , raf Kinases/metabolism
2.
Cell ; 140(4): 517-28, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20178744

ABSTRACT

Despite its toxicity, H(2)O(2) is produced as a signaling molecule that oxidizes critical cysteine residues of effectors such as protein tyrosine phosphatases in response to activation of cell surface receptors. It has remained unclear, however, how H(2)O(2) concentrations above the threshold required to modify effectors are achieved in the presence of the abundant detoxification enzymes peroxiredoxin (Prx) I and II. We now show that PrxI associated with membranes is transiently phosphorylated on tyrosine-194 and thereby inactivated both in cells stimulated via growth factor or immune receptors in vitro and in those at the margin of healing cutaneous wounds in mice. The localized inactivation of PrxI allows for the transient accumulation of H(2)O(2) around membranes, where signaling components are concentrated, while preventing the toxic accumulation of H(2)O(2) elsewhere. In contrast, PrxII was inactivated not by phosphorylation but rather by hyperoxidation of its catalytic cysteine during sustained oxidative stress.


Subject(s)
Hydrogen Peroxide/metabolism , Peroxiredoxins/metabolism , Animals , Cell Membrane/metabolism , Enzyme Activation , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred BALB C , Models, Biological , NIH 3T3 Cells , Oxidative Stress , Rats , Receptor Protein-Tyrosine Kinases/metabolism , Skin/metabolism , Tyrosine/metabolism , Wound Healing
3.
J Cell Sci ; 133(20)2020 10 21.
Article in English | MEDLINE | ID: mdl-32938684

ABSTRACT

PTPRT has been known to regulate synaptic formation and dendritic arborization of hippocampal neurons. PTPRT-/- null and PTPRT-D401A mutant mice displayed enhanced depression-like behaviors compared with wild-type mice. Transient knockdown of PTPRT in the dentate gyrus enhanced the depression-like behaviors of wild-type mice, whereas rescued expression of PTPRT ameliorated the behaviors of PTPRT-null mice. Chronic stress exposure reduced expression of PTPRT in the hippocampus of mice. In PTPRT-deficient mice the expression of GluR2 (also known as GRIA2) was attenuated as a consequence of dysregulated tyrosine phosphorylation, and the long-term potentiation at perforant-dentate gyrus synapses was augmented. The inhibitory synaptic transmission of the dentate gyrus and hippocampal GABA concentration were reduced in PTPRT-deficient mice. In addition, the hippocampal expression of GABA transporter GAT3 (also known as SLC6A11) was decreased, and its tyrosine phosphorylation was increased in PTPRT-deficient mice. PTPRT-deficient mice displayed reduced numbers and neurite length of newborn granule cells in the dentate gyrus and had attenuated neurogenic ability of embryonic hippocampal neural stem cells. In conclusion, our findings show that the physiological roles of PTPRT in hippocampal neurogenesis, as well as synaptic functions, are involved in the pathogenesis of depressive disorder.


Subject(s)
Depression , Neurogenesis , Animals , Dentate Gyrus , Hippocampus , Mice , Mice, Knockout , Neurogenesis/genetics , Neurons , Synapses
4.
Mol Psychiatry ; 26(8): 3737-3750, 2021 08.
Article in English | MEDLINE | ID: mdl-32989244

ABSTRACT

Guanine nucleotide binding protein (G protein) gamma 8 (Gng8) is a subunit of G proteins and expressed in the medial habenula (MHb) and interpeduncular nucleus (IPN). Recent studies have demonstrated that Gng8 is involved in brain development; however, the roles of Gng8 on cognitive function have not yet been addressed. In the present study, we investigated the expression of Gng8 in the brain and found that Gng8 was predominantly expressed in the MHb-IPN circuit of the mouse brain. We generated Gng8 knockout (KO) mice by CRISPR/Cas9 system in order to assess the role of Gng8 on cognitive function. Gng8 KO mice exhibited deficiency in learning and memory in passive avoidance and Morris water maze tests. In addition, Gng8 KO mice significantly reduced long-term potentiation (LTP) in the hippocampus compared to that of wild-type (WT) mice. Furthermore, we observed that levels of acetylcholine (ACh) and choline acetyltransferase (ChAT) in the MHb and IPN of Gng8 KO mice were significantly decreased, compared to WT mice. The administration of nAChR α4ß2 agonist A85380 rescued memory impairment in the Gng8 KO mice, suggesting that Gng8 regulates cognitive function via modulation of cholinergic activity. Taken together, Gng8 is a potential therapeutic target for memory-related diseases and/or neurodevelopmental diseases.


Subject(s)
Habenula , Acetylcholine , Animals , Learning , Maze Learning , Mice , Mice, Knockout , Nicotinic Agonists
5.
Cell Mol Life Sci ; 76(13): 2647-2662, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30903204

ABSTRACT

Hepatitis B virus (HBV) X protein (HBx) is associated with hepatocarcinogenesis. E2-EPF ubiquitin carrier protein (UCP) catalyzes ubiquitination of itself and von Hippel-Lindau protein (pVHL) for degradation and associates with tumor growth and metastasis. However, it remains unknown whether HBx modulates the enzyme activity of UCP and thereby influences hepatocarcinogenesis. Here, we show that UCP is highly expressed in liver tissues of HBx-transgenic mice, but not non-transgenic mice. UCP was more frequently expressed in HBV-positive liver cancers than in HBV-negative liver cancers. HBx binds to UCP specifically and serotype independently, and forms a ternary complex with UCP and pVHL. HBx inhibits self-ubiquitination of UCP, but enhances UCP-mediated pVHL ubiquitination, resulting in stabilization of hypoxia-inducible factor-1α and -2α. HBx and UCP stabilize each other by mutually inhibiting their ubiquitination. HBx promotes cellular proliferation and metastasis via UCP. Our findings suggest that UCP plays a key role in HBV-related hepatocarcinogenesis.


Subject(s)
Carcinoma, Hepatocellular/secondary , Hepatitis B/complications , Liver Neoplasms/pathology , Trans-Activators/metabolism , Ubiquitin-Conjugating Enzymes/chemistry , Animals , Apoptosis , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/virology , Cell Proliferation , Disease Progression , Female , Hepatitis B/virology , Hepatitis B virus/isolation & purification , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/virology , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Protein Stability , Signal Transduction , Trans-Activators/genetics , Tumor Cells, Cultured , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination , Viral Regulatory and Accessory Proteins , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Xenograft Model Antitumor Assays
6.
Nucleic Acids Res ; 46(17): 8832-8847, 2018 09 28.
Article in English | MEDLINE | ID: mdl-29992318

ABSTRACT

Genomic sequencing of hepatocellular carcinoma (HCC) uncovers a paucity of actionable mutations, underscoring the necessity to exploit epigenetic vulnerabilities for therapeutics. In HCC, EZH2-mediated H3K27me3 represents a major oncogenic chromatin modification, but how it modulates the therapeutic vulnerability of signaling pathways remains unknown. Here, we show EZH2 acts antagonistically to AKT signaling in maintaining H3K27 methylome through epigenetic silencing of IGFBP4. ChIP-seq revealed enrichment of Ezh2/H3K27me3 at silenced loci in HBx-transgenic mouse-derived HCCs, including Igfbp4 whose down-regulation significantly correlated with EZH2 overexpression and poor survivals of HCC patients. Functional characterizations demonstrated potent growth- and invasion-suppressive functions of IGFBP4, which was associated with transcriptomic alterations leading to deregulation of multiple signaling pathways. Mechanistically, IGFBP4 stimulated AKT/EZH2 phosphorylation to abrogate H3K27me3-mediated silencing, forming a reciprocal feedback loop that suppressed core transcription factor networks (FOXA1/HNF1A/HNF4A/KLF9/NR1H4) for normal liver homeostasis. Consequently, the in vivo tumorigenicity of IGFBP4-silenced HCC cells was vulnerable to pharmacological inhibition of EZH2, but not AKT. Our study unveils chromatin regulation of a novel liver tumor suppressor IGFBP4, which constitutes an AKT-EZH2 reciprocal loop in driving H3K27me3-mediated epigenetic reprogramming. Defining the aberrant chromatin landscape of HCC sheds light into the mechanistic basis of effective EZH2-targeted inhibition.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Hepatocellular/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Histone Code/genetics , Histones/metabolism , Insulin-Like Growth Factor Binding Protein 4/deficiency , Liver Neoplasms/genetics , Tumor Suppressor Proteins/deficiency , Animals , Cell Line, Tumor , Chromatin Immunoprecipitation , Female , Humans , Insulin-Like Growth Factor Binding Protein 4/genetics , Insulin-Like Growth Factor Binding Protein 4/physiology , Liver Neoplasms, Experimental/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Molecular Targeted Therapy , Prognosis , Protein Interaction Mapping , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , RNA, Neoplasm/genetics , Sequence Analysis, RNA , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , Xenograft Model Antitumor Assays
7.
Mol Reprod Dev ; 86(11): 1628-1638, 2019 11.
Article in English | MEDLINE | ID: mdl-31373105

ABSTRACT

Understanding preimplantation embryo development has important implications for assisted reproductive technologies (ARTs) after the introduction of in vitro fertilisation and embryo transfer because most embryonic losses occur during pre/peri-implantation. Recent studies have shown that tight junctions (TJs) are important components for embryos to develop to the blastocyst stage. However, their biological function after cavitation has not been extensively studied. We examined TJ assembly focusing on coxsackievirus and adenovirus receptor (Cxadr) and A disintegrin and metalloproteinase 10 (Adam10) using siRNA and/or an Adam10-specific inhibitor (GI254023X). TJ-associated genes, including occludin and tight junction protein 1 (Tjp1), were downregulated in the Cxadr knockdown (KD) embryos but were unaltered in Adam10 KD embryos. However, Adam10 KD or chemical inhibition affected subcellular localisation of Adam10, Cxadr, and Tjp1, leading to disrupted TJ assembly. Furthermore, Cxadr KD or GI254023X-treated blastocysts showed a relatively smaller outgrowth area and aberrant expression of transcription factor AP-2γ, a trophoblast-specific marker in the in vitro embryo outgrowth assay. In summary, we demonstrated that the Cxadr-Adam10 complex might moderate TJ integrity/stability and play pivotal roles during early embryonic development. Collectively, understanding the establishment of the TJ complex and its integrity will provide insight into translational research for predicting and selecting developmental competency for ART.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Membrane Proteins/metabolism , Multiprotein Complexes/metabolism , Tight Junctions/metabolism , Trophoblasts/metabolism , ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/genetics , Animals , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Down-Regulation , Membrane Proteins/genetics , Mice , Multiprotein Complexes/genetics , RNA, Small Interfering/genetics , Tight Junctions/genetics , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism , Trophoblasts/cytology , Zonula Occludens-1 Protein/biosynthesis , Zonula Occludens-1 Protein/genetics
8.
Mol Cell ; 44(4): 545-58, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-22099303

ABSTRACT

Cellular antioxidant enzymes play crucial roles in aerobic organisms by eliminating detrimental oxidants and maintaining the intracellular redox homeostasis. Therefore, the function of antioxidant enzymes is inextricably linked to the redox-dependent activities of multiple proteins and signaling pathways. Here, we report that the VEGFR2 RTK has an oxidation-sensitive cysteine residue whose reduced state is preserved specifically by peroxiredoxin II (PrxII) in vascular endothelial cells. In the absence of PrxII, the cellular H(2)O(2) level is markedly increased and the VEGFR2 becomes inactive, no longer responding to VEGF stimulation. Such VEGFR2 inactivation is due to the formation of intramolecular disulfide linkage between Cys1199 and Cys1206 in the C-terminal tail. Interestingly, the PrxII-mediated VEGFR2 protection is achieved by association of two proteins in the caveolae. Furthermore, PrxII deficiency suppresses tumor angiogenesis in vivo. This study thus demonstrates a physiological function of PrxII as the residential antioxidant safeguard specific to the redox-sensitive VEGFR2.


Subject(s)
Antioxidants/metabolism , Aorta/enzymology , Endothelial Cells/enzymology , Endothelium, Vascular/enzymology , Neovascularization, Pathologic/enzymology , Peroxiredoxins , Vascular Endothelial Growth Factor Receptor-2 , Animals , Aorta/cytology , Carcinoma, Lewis Lung/enzymology , Carcinoma, Lewis Lung/pathology , Caveolae/enzymology , Cysteine/chemistry , Cysteine/metabolism , Disulfides/chemistry , Disulfides/metabolism , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Gene Silencing , Humans , Hydrogen Peroxide/metabolism , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , Oxidation-Reduction , Peroxiredoxins/antagonists & inhibitors , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
9.
Stem Cells ; 34(5): 1188-97, 2016 05.
Article in English | MEDLINE | ID: mdl-26866938

ABSTRACT

Redox regulation in cancer stem cells (CSCs) is viewed as a good target for cancer therapy because redox status plays an important role in cancer stem-cell maintenance. Here, we investigated the role of Peroxiredoxin II (Prx II), an antioxidant enzyme, in association with maintenance of liver CSCs. Our study demonstrates that Prx II overexpressed in liver cancer cells has high potential for self-renewal activity. Prx II expression significantly corelated with expression of epithelial-cell adhesion molecules (EpCAM) and cytokerain 19 in liver cancer tissues of hepatocellular carcinoma (HCC) patients. Downregulation of Prx II in Huh7 cells with treatment of siRNA reduced expression of EpCAM and CD133 as well as Sox2 in accordance with increased ROS and apoptosis, which were reversed in Huh7-hPrx II cells. Huh7-hPrx II cells exhibited strong sphere-formation activity compared with mock cells. Vascular endothelial growth factor (VEGF) exposure enhanced sphere formation, cell-surface expression of EpCAM and CD133, and pSTAT3 along with activation of VEGF receptor 2 in Huh7-hPrx II cells. The result also emerged in Huh7-H-ras(G12V) and SK-HEP-1-H-ras(G12V) cells with high-level expression of Prx II. Prx II was involved in regulation of VEGF driving cancer stem cells through VEGFR-2/STAT3 signaling to upregulate Bmi1 and Sox2. In addition, knockdown of Prx II in Huh7-H-ras(G12V) cells showed significant reduction in cell migration in vitro and in tumorigenic potential in vivo. Taken together, all the results demonstrated that Prx II plays a key role in the CSC self-renewal of HCC cells through redox regulation. Stem Cells 2016;34:1188-1197.


Subject(s)
Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Peroxiredoxins/metabolism , Animals , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Self Renewal/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/drug effects , Oxidation-Reduction/drug effects , Proto-Oncogene Proteins p21(ras)/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
10.
J Pathol ; 238(5): 651-64, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26800240

ABSTRACT

Enhancer of zeste homolog 2 (EZH2) catalyses histone H3 lysine 27 trimethylation (H3K27me3) to silence tumour-suppressor genes in hepatocellular carcinoma (HCC) but the process of locus-specific recruitment remains elusive. Here we investigated the transcription factors involved and the molecular consequences in HCC development. The genome-wide distribution of H3K27me3 was determined by chromatin immunoprecipitation coupled with high-throughput sequencing or promoter array analyses in HCC cells from hepatitis B virus (HBV) X protein transgenic mouse and human cell models. Transcription factor binding site analysis was performed to identify EZH2-interacting transcription factors followed by functional characterization. Our cross-species integrative analysis revealed a crucial link between Yin Yang 1 (YY1) and EZH2-mediated H3K27me3 in HCC. Gene expression analysis of human HBV-associated HCC specimens demonstrated concordant overexpression of YY1 and EZH2, which correlated with poor survival of patients in advanced stages. The YY1 binding motif was significantly enriched in both in vivo and in vitro H3K27me3-occupied genes, including genes for 15 tumour-suppressive microRNAs. Knockdown of YY1 reduced not only global H3K27me3 levels, but also EZH2 and H3K27me3 promoter occupancy and DNA methylation, leading to the transcriptional up-regulation of microRNA-9 isoforms in HCC cells. Concurrent EZH2 knockdown and 5-aza-2'-deoxycytidine treatment synergistically increased the levels of microRNA-9, which reduced the expression and transcriptional activity of nuclear factor-κB (NF-κB). Functionally, YY1 promoted HCC tumourigenicity and inhibited apoptosis of HCC cells, at least partially through NF-κB activation. In conclusion, YY1 overexpression contributes to EZH2 recruitment for H3K27me3-mediated silencing of tumour-suppressive microRNAs, thereby activating NF-κB signalling in hepatocarcinogenesis.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Gene Silencing , Liver Neoplasms/metabolism , MicroRNAs/metabolism , NF-kappa B/metabolism , YY1 Transcription Factor/metabolism , Animals , Apoptosis , Binding Sites , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Cell Proliferation , DNA Methylation , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/virology , Lysine , Methylation , Mice, Nude , Mice, Transgenic , MicroRNAs/genetics , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic , RNA Interference , Signal Transduction , Time Factors , Trans-Activators/genetics , Trans-Activators/metabolism , Transfection , Tumor Burden , Up-Regulation , Viral Regulatory and Accessory Proteins , YY1 Transcription Factor/genetics
11.
J Biol Chem ; 290(18): 11432-42, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25802339

ABSTRACT

Collagen-induced platelet signaling is mediated by binding to the primary receptor glycoprotein VI (GPVI). Reactive oxygen species produced in response to collagen have been found to be responsible for the propagation of GPVI signaling pathways in platelets. Therefore, it has been suggested that antioxidant enzymes could down-regulate GPVI-stimulated platelet activation. Although the antioxidant enzyme peroxiredoxin II (PrxII) has emerged as having a role in negatively regulating signaling through various receptors by eliminating H2O2 generated upon receptor stimulation, the function of PrxII in collagen-stimulated platelets is not known. We tested the hypothesis that PrxII negatively regulates collagen-stimulated platelet activation. We analyzed PrxII-deficient murine platelets. PrxII deficiency enhanced GPVI-mediated platelet activation through the defective elimination of H2O2 and the impaired protection of SH2 domain-containing tyrosine phosphatase 2 (SHP-2) against oxidative inactivation, which resulted in increased tyrosine phosphorylation of key components for the GPVI signaling cascade, including Syk, Btk, and phospholipase Cγ2. Interestingly, PrxII-mediated antioxidative protection of SHP-2 appeared to occur in the lipid rafts. PrxII-deficient platelets exhibited increased adhesion and aggregation upon collagen stimulation. Furthermore, in vivo experiments demonstrated that PrxII deficiency facilitated platelet-dependent thrombus formation in injured carotid arteries. This study reveals that PrxII functions as a protective antioxidant enzyme against collagen-stimulated platelet activation and platelet-dependent thrombosis.


Subject(s)
Antioxidants/metabolism , Blood Platelets/drug effects , Blood Platelets/physiology , Collagen/pharmacology , Peroxiredoxins/metabolism , Platelet Membrane Glycoproteins/metabolism , Animals , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/physiopathology , Hydrogen Peroxide/metabolism , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mice , Peroxiredoxins/deficiency , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Thrombosis/metabolism , Thrombosis/physiopathology , Tyrosine/metabolism
12.
BMC Complement Altern Med ; 16: 42, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26832364

ABSTRACT

BACKGROUND: Cervical cancer is the second most common cancer in females. Recent reports have revealed the critical role of cervical cancer stem cells (CSCs) in tumorigenicity and metastasis. Previously we demonstrated that A1E exerts an anti-proliferative action, which inhibits the growth of cervical cancer cells. METHODS: A1E is composed of 11 oriental medicinal herbs. Cervical cancer cell culture, wund healing and invasion assay, flow cytometry, sheroid formation assay, and wstern blot assays were performed in HPV 16-positive SiHa cell and HPV 16-negative C33A cells. RESULTS: A1E targets the E6 and E7 oncogenes; thus, A1E significantly inhibited proliferation of human papilloma virus (HPV) 16-positive SiHa cells, it did not inhibit the proliferation of HPV-negative C33A cells. Accordingly, we investigated whether A1E can regulate epithelial-to-mesenchymal transition (EMT), CSC self-renewal, and stemness-related gene expression in cervical cancer cells. Down rgulation of cell migration, cell invasion, and EMT was observed in A1E-treated SiHa cells. Specifically, A1E-treated SiHa cells showed significant decreases in OCT-3/4 and Sox2 expression levels and in sphere formation. Moreover, CSCs makers ALDH+ and ALDH, CD133 double positive cell were significantly decreased in A1E-treated SiHa cells. However, A1E treatment did not down regulate ALDH+ expression and the number of ALDH/CD133 double positive cells in C33A cells. CONCLUSIONS: Taken together, A1E can inhibit CSCs and reduce the expression of stemness markers. Treating CSCs with A1E may be a potential therapy for cervical cancer.


Subject(s)
Human papillomavirus 16/drug effects , Neoplastic Stem Cells/drug effects , Papillomavirus Infections/drug therapy , Plant Extracts/therapeutic use , Uterine Cervical Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Plants, Medicinal/chemistry
13.
J Hepatol ; 63(2): 429-36, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25828473

ABSTRACT

BACKGROUND & AIMS: Overexpression of FoxM1 correlates with poor prognosis in hepatocellular carcinoma (HCC). Moreover, the Ras-signaling pathway is found to be ubiquitously activated in HCC through epigenetic silencing of the Ras-regulators. We investigated the roles of FoxM1 in Ras-driven HCC, and on HCC cells with stem-like features. METHODS: We employed a transgenic mouse model that expresses the oncogenic Ras in the liver. That strain was crossed with a strain that harbor floxed alleles of FoxM1 and the MxCre gene that allows conditional deletion of FoxM1. FoxM1 alleles were deleted after development of HCC, and the effects on the tumors were analyzed. Also, FoxM1 siRNA was used in human HCC cell lines to determine its role in the survival of the HCC cells with stem cell features. RESULTS: Ras-driven tumors overexpress FoxM1. Deletion of FoxM1 inhibits HCC progression. There was increased accumulation of reactive oxygen species (ROS) in the FoxM1 deleted HCC cells. Moreover, FoxM1 deletion caused a disproportionate loss of the CD44+ and EpCAM+ HCC cells in the tumors. We show that FoxM1 directly activates expression of CD44 in human HCC cells. Moreover, the human HCC cells with stem cell features are addicted to FoxM1 for ROS-regulation and survival. CONCLUSION: Our results provide genetic evidence for an essential role of FoxM1 in the progression of Ras-driven HCC. In addition, FoxM1 is required for the expression of CD44 in HCC cells. Moreover, FoxM1 plays a critical role in the survival of the HCC cells with stem cell features by regulating ROS.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Hepatocellular/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Stem Cells/pathology , ras Proteins/genetics , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Disease Progression , Forkhead Box Protein M1 , Forkhead Transcription Factors/biosynthesis , Humans , Immunohistochemistry , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Prognosis , Signal Transduction , Stem Cells/metabolism , ras Proteins/biosynthesis
14.
Biochem Biophys Res Commun ; 466(4): 676-81, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26392315

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common malignancies and chronic hepatitis B virus (HBV) infection is a major risk factor for HCC. Hepatitis B virus X (HBx) protein relates to trigger oncogenesis. HBx has oncogenic properties with a hyperproliferative response to HCC. Nuclear protein 1 (NUPR1) is a stress-response protein, frequently upregulated in several cancers. Recent data revealed that NUPR1 is involved in tumor progression, but its function in HCC is not revealed yet. Here we report HBx can induce NUPR1 in patients, mice, and HCC cell lines. In an HBx transgenic mouse model, we found that HBx overexpression upregulates NUPR1 expression consistently with tumor progression. Further, in cultured HBV positive cells, HBx knockdown induces downregulation of NUPR1. Smad4 is a representative transcription factor, regulated by HBx, and we showed that HBx upregulates NUPR1 by Smad4 dependent way. We found that NUPR1 can inhibit cell death and induce vasculogenic mimicry in HCC cell lines. Moreover, NUPR1 silencing in HepG2-HBx showed reduced cell motility. These results suggest that HBx can modulate NUPR1 expression through the Smad4 pathway and NUPR1 has a role in hepatocellular carcinoma progression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Hepatocellular/etiology , DNA-Binding Proteins/metabolism , Hepatitis B virus/pathogenicity , Liver Neoplasms/etiology , Neoplasm Proteins/metabolism , Trans-Activators/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , Hep G2 Cells , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Hepatitis B, Chronic/complications , Host-Pathogen Interactions , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Signal Transduction , Smad4 Protein/metabolism , Trans-Activators/genetics , Viral Regulatory and Accessory Proteins
15.
Stem Cells ; 32(4): 998-1011, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24715692

ABSTRACT

Redox balance has been suggested as an important determinant of "stemness" in embryonic stem cells (ESCs). In this study, we demonstrate that peroxiredoxin (Prx) plays a pivotal role in maintenance of ESC stemness during neurogenesis through suppression of reactive oxygen species (ROS)-sensitive signaling. During neurogenesis, Prx I and Oct4 are expressed in a mutually dependent manner and their expression is abruptly downregulated by an excess of ROS. Thus, in Prx I(-/-) or Prx II(-/-) ESCs, rapid loss of stemness can occur due to spontaneous ROS overload, leading to their active commitment into neurons; however, stemness is restored by the addition of an antioxidant or an inhibitor of c-Jun N-terminal kinase (JNK). In addition, Prx I and Prx II appear to have a tight association with the mechanism underlying the protection of ESC stemness in developing teratomas. These results suggest that Prx functions as a protector of ESC stemness by opposing ROS/JNK cascades during neurogenesis. Therefore, our findings have important implications for understanding of maintenance of ESC stemness through involvement of antioxidant enzymes and may lead to development of an alternative stem cell-based therapeutic strategy for production of high-quality neurons in large quantity.


Subject(s)
Embryonic Stem Cells/enzymology , MAP Kinase Kinase 4/metabolism , Neurogenesis/physiology , Peroxiredoxins/metabolism , Reactive Oxygen Species/metabolism , Animals , Embryonic Stem Cells/cytology , MAP Kinase Kinase 4/genetics , Mice , Mice, Knockout , Peroxiredoxins/genetics
16.
J Immunol ; 191(8): 4029-37, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24048895

ABSTRACT

Peroxiredoxin (Prx) II is an intracellular antioxidant molecule that eliminates hydrogen peroxide, employing a high substrate-binding affinity. PrxII deficiency increases the levels of intracellular reactive oxygen species in many types of cells, which may increase reactive oxygen species-mediated inflammation. In this study, we investigated the susceptibility of PrxII knockout (KO) mice to experimentally induced colitis and the effects of PrxII on the immune system. Wild-type mice displayed pronounced weight loss, high mortality, and colon shortening after dextran sulfate sodium administration, whereas colonic inflammation was significantly attenuated in PrxII KO mice. Although macrophages were hyperactivated in PrxII KO mice, the amount of IFN-γ and IL-17 produced by CD4(+) T cells was substantially reduced. Foxp3(+) regulatory T (Treg) cells were elevated, and Foxp3 protein expression was increased in the absence of PrxII in vitro and in vivo. Restoration of PrxII into KO cells suppressed the increased Foxp3 expression. Interestingly, endogenous PrxII was inactivated through hyperoxidation during Treg cell development. Furthermore, PrxII deficiency stabilized FoxO1 expression by reducing mouse double minute 2 homolog expression and subsequently activated FoxO1-mediated Foxp3 gene transcription. PrxII overexpression, in contrast, reduced FoxO1 and Foxp3 expression. More interestingly, adoptive transfer of naive CD4(+) T cells from PrxII KO mice into immune-deficient mice attenuated T cell-induced colitis, with a reduction in mouse double minute 2 homolog expression and an increase in FoxO1 and Foxp3 expression. These results suggest that inactivation of PrxII is important for the stability of FoxO1 protein, which subsequently mediates Foxp3(+) Treg cell development, thereby attenuating colonic inflammation.


Subject(s)
Colitis/immunology , Forkhead Transcription Factors/metabolism , Peroxiredoxins/metabolism , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Colitis/genetics , Dextran Sulfate , Forkhead Box Protein O1 , Interferon-gamma/metabolism , Interleukin-17/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peroxiredoxins/genetics , Proto-Oncogene Proteins c-mdm2/biosynthesis , Proto-Oncogene Proteins c-mdm2/metabolism , Reactive Oxygen Species/metabolism , T-Lymphocytes, Regulatory/metabolism
17.
Gut ; 63(11): 1793-804, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24440987

ABSTRACT

BACKGROUND: Androgen receptor (AR) signalling contributes to male predominance in hepatocellular carcinoma (HCC), which is more pronounced in HBV-endemic areas. Cell cycle-related kinase (CCRK) is essential for AR-induced hepatocarcinogenesis but its molecular function in HBV-associated HCC remains obscure. OBJECTIVE: To determine the molecular function of CCRK in HBV-associated HCC. DESIGN: Transcriptional regulation was assessed by chromatin immunoprecipitation, promoter mutation and luciferase reporter assays. Hepatocellular proliferation and tumourigenesis were examined by colony formation, soft agar assays and using HBV X protein (HBx) transgenic mice with low-dose exposure to diethylnitrosamine. Protein expressions were examined in clinical samples and correlated with patient survival by log-rank Mantel-Cox test. RESULTS: Overexpression of CCRK, but not its kinase-defective mutant, activated ß-catenin/T cell factor signalling through phosphorylation of glycogen synthase kinase-3ß (GSK-3ß) at Ser9, led to upregulation of AR transcriptional activity and, subsequently, expression of HBx. The viral transactivator in turn induced CCRK expression through enhanced AR signalling, thus forming a positive regulatory loop. RNA interference silencing of CCRK, which suppressed the CCRK/GSK-3ß/ß-catenin/AR regulatory loop, significantly suppressed HBx-induced hepatocellular proliferation (p=0.001) and transformation (p<0.001) and remarkably reduced >80% diethylnitrosamine-mediated hepatocarcinogenesis in HBx transgenic mice. Finally, patients with HBV-associated HCC with concordant overexpression of CCRK, GSK-3ß phosphorylation at Ser9, active dephosphorylated ß-catenin and AR phosphorylation at Ser81 had poorer overall (HR=31.26, p<0.0001) and disease-free (HR=3.60, p<0.01) survival rates. CONCLUSIONS: Our findings highlight the critical role of CCRK in a self-reinforcing circuitry that regulates HBV-associated hepatocarcinogenesis. Further characterisation of this intricate viral-host signalling may provide new prognostic biomarkers and therapeutic targets for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cyclin-Dependent Kinases/biosynthesis , Hepatitis B/complications , Liver Neoplasms/metabolism , Carcinogenesis , Carcinoma, Hepatocellular/virology , Cells, Cultured , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Host-Pathogen Interactions , Humans , Liver Neoplasms/virology , Prognosis , Receptors, Androgen/metabolism , TCF Transcription Factors/metabolism , Trans-Activators/metabolism , Transcriptional Activation , Up-Regulation , Viral Regulatory and Accessory Proteins , beta Catenin/metabolism , Cyclin-Dependent Kinase-Activating Kinase
18.
Mol Cancer ; 13: 222, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25255877

ABSTRACT

BACKGROUND: The metastasis of hematogenous cancer cells is associated with abnormal glycosylation such as sialyl lewis antigens. Although the hepatitis B virus X protein (HBx) plays important role in liver disease, the precise function of HBx on aberrant glycosylation for metastasis remains unclear. METHODS: The human hepatocellular carcinoma tissues, HBx transgenic mice and HBx-transfected cells were used to check the correlation of expressions between HBx and Sialyl lewis antigen for cancer metastasis. To investigate whether expression levels of glycosyltransferases induced in HBx-transfected cells are specifically associated with sialyl lewis A (SLA) synthesis, which enhances metastasis by interaction of liver cancer cells with endothelial cells, ShRNA and siRNAs targeting specific glycosyltransferases were used. RESULTS: HBx expression in liver cancer region of HCC is associated with the specific synthesis of SLA. Furthermore, the SLA was specifically induced both in liver tissues from HBx-transgenic mice and in in vitro HBx-transfected cells. HBx increased transcription levels and activities of α2-3 sialyltransferases (ST3Gal III), α1-3/4 fucosyltransferases III and VII (FUT III and VII) genes, which were specific for SLA synthesis, allowing dramatic cell-cell adhesion for metastatic potential. Interestingly, HBx specifically induced expression of N-acetylglucosamine-ß1-3 galactosyltransferase V (ß1-3GalT 5) gene associated with the initial synthesis of sialyl lewis A, but not ß1-4GalT I. The ß1-3GalT 5 shRNA suppressed SLA expression by HBx, blocking the adhesion of HBx-transfected cells to the endothelial cells. Moreover, ß1-3GalT 5 silencing suppressed lung metastasis of HBx-transfected cells in in vivo lung metastasis system. CONCLUSION: HBx targets the specific glycosyltransferases for the SLA synthesis and this process regulates hematogenous cancer cell adhesion to endothelial cells for cancer metastasis.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Glycosyltransferases/metabolism , Liver Neoplasms/metabolism , Liver/metabolism , Liver/virology , Oligosaccharides/metabolism , Trans-Activators/metabolism , Adult , Animals , CA-19-9 Antigen , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Glycosylation , Hepatitis B virus/physiology , Human Umbilical Vein Endothelial Cells , Humans , Lewis X Antigen/metabolism , Liver/pathology , Liver Neoplasms/pathology , Liver Neoplasms/virology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Neoplasm Metastasis/pathology , Sialyl Lewis X Antigen , Viral Regulatory and Accessory Proteins
19.
Blood ; 119(10): 2346-57, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-22207736

ABSTRACT

With the use of ChIP on microarray assays in primary leukemia samples, we report that acute myeloid leukemia (AML) blasts exhibit significant alterations in histone H3 acetylation (H3Ac) levels at > 1000 genomic loci compared with CD34(+) progenitor cells. Importantly, core promoter regions tended to have lower H3Ac levels in AML compared with progenitor cells, which suggested that a large number of genes are epigenetically silenced in AML. Intriguingly, we identified peroxiredoxin 2 (PRDX2) as a novel potential tumor suppressor gene in AML. H3Ac was decreased at the PRDX2 gene promoter in AML, which correlated with low mRNA and protein expression. We also observed DNA hypermethylation at the PRDX2 promoter in AML. Low protein expression of the antioxidant PRDX2 gene was clinically associated with poor prognosis in patients with AML. Functionally, PRDX2 acted as inhibitor of myeloid cell growth by reducing levels of reactive oxygen species (ROS) generated in response to cytokines. Forced PRDX2 expression inhibited c-Myc-induced leukemogenesis in vivo on BM transplantation in mice. Taken together, epigenome-wide analyses of H3Ac in AML led to the identification of PRDX2 as an epigenetically silenced growth suppressor, suggesting a possible role of ROS in the malignant phenotype in AML.


Subject(s)
DNA Methylation , Histones/metabolism , Peroxiredoxins/genetics , Tumor Suppressor Proteins/genetics , Acetylation , Acute Disease , Adolescent , Adult , Aged , Aged, 80 and over , Cell Proliferation , Cells, Cultured , Epigenesis, Genetic , Female , Gene Expression Profiling/methods , Genome-Wide Association Study , HL-60 Cells , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , Peroxiredoxins/metabolism , Reactive Oxygen Species/metabolism , Tumor Suppressor Proteins/metabolism , U937 Cells , Young Adult
20.
Int J Mol Sci ; 15(3): 4126-41, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24608925

ABSTRACT

Hepatitis C virus (HCV) has become a major public health issue. It is prevalent in most countries. HCV infection frequently begins without clinical symptoms, before progressing to persistent viremia, chronic hepatitis, cirrhosis and hepatocellular carcinoma (HCC) in the majority of patients (70% to 80%). Alcohol is an independent cofactor that accelerates the development of HCC in chronic hepatitis C patients. The purpose of the current study was to evaluate ethanol-induced hepatic changes in HCV core-Tg mice and mutant core Tg mice. Wild type (NTG), core wild-Tg mice (TG-K), mutant core 116-Tg mice (TG-116) and mutant core 99-Tg mice (TG-99) were used in this investigation. All groups were given drinking water with 10% ethanol and 5% sucrose for 13 weeks. To observe liver morphological changes, we performed histopathological and immunohistochemical examinations. Histopathologically, NTG, TG-K and TG-116 mice showed moderate centrilobular necrosis, while severe centrilobular necrosis and hepatocyte dissociation were observed in TG-99 mice with increasing lymphocyte infiltration and piecemeal necrosis. In all groups, a small amount of collagen fiber was found, principally in portal areas. None of the mice were found to have myofibroblasts based on immunohistochemical staining specific for α-SMA. CYP2E1-positive cells were clearly detected in the centrilobular area in all groups. In the TG-99 mice, we also observed cells positive for CK8/18, TGF-ß1 and phosphorylated (p)-Smad2/3 and p21 around the necrotic hepatocytes in the centrilobular area (p < 0.01). Based on our data, alcohol intake induced piecemeal necrosis and hepatocyte dissociation in the TG-99 mice. These phenomena involved activation of the TGF-ß1/p-Smad2/3/p21 signaling pathway in hepatocytes. Data from this study will be useful for elucidating the association between alcohol intake and HCV infection.


Subject(s)
Ethanol/toxicity , Liver/drug effects , Mutation , Viral Core Proteins/genetics , Actins/metabolism , Animals , Central Nervous System Depressants/toxicity , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytochrome P-450 CYP2E1/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Immunohistochemistry , Keratin-18/metabolism , Keratin-8/metabolism , Liver/metabolism , Liver/pathology , Liver Diseases/etiology , Liver Diseases/genetics , Liver Diseases/metabolism , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Muscle, Smooth/chemistry , Necrosis/chemically induced , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Viral Core Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL