Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Nucleic Acids Res ; 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38966995

ABSTRACT

Nuclear actin-based movements support DNA double-strand break (DSB) repair. However, molecular determinants that promote filamentous actin (F-actin) formation on the damaged chromatin remain undefined. Here we describe the DYRK1A kinase as a nuclear activity that promotes local F-actin assembly to support DSB mobility and repair, accomplished in part by its targeting of actin nucleator spire homolog 1 (Spir1). Indeed, perturbing DYRK1A-dependent phosphorylation of S482 mis-regulated Spir1 accumulation at damaged-modified chromatin, and led to compromised DSB-associated actin polymerization and attenuated DNA repair. Our findings uncover a role of the DYRK1A-Spir1 axis in nuclear actin dynamics during early DSB responses, and highlight the intricate details of nuclear cytoskeletal network in DSB repair and genome stability maintenance.

2.
J Pathol ; 259(2): 163-179, 2023 02.
Article in English | MEDLINE | ID: mdl-36420735

ABSTRACT

Invadopodia are actin-rich membrane protrusions that digest the matrix barrier during cancer metastasis. Since the discovery of invadopodia, they have been visualized as localized and dot-like structures in different types of cancer cells on top of a 2D matrix. In this investigation of Epstein-Barr virus (EBV)-associated nasopharyngeal carcinoma (NPC), a highly invasive cancer frequently accompanied by neck lymph node and distal organ metastases, we revealed a new form of invadopodium with mobilizing features. Integration of live-cell imaging and molecular assays revealed the interaction of macrophage-released TNFα and EBV-encoded latent membrane protein 1 (LMP1) in co-activating the EGFR/Src/ERK/cortactin and Cdc42/N-WASP signaling axes for mobilizing the invadopodia with lateral movements. This phenomenon endows the invadopodia with massive degradative power, visualized as a shift of focal dot-like digestion patterns on a 2D gelatin to a dendrite-like digestion pattern. Notably, single stimulation of either LMP1 or TNFα could only enhance the number of ordinary dot-like invadopodia, suggesting that the EBV infection sensitizes the NPC cells to form mobilizing invadopodia when encountering a TNFα-rich tumor microenvironment. This study unveils the interplay of EBV and stromal components in driving the invasive potential of NPC via unleashing the propulsion of invadopodia in overcoming matrix hurdles. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Epstein-Barr Virus Infections , Nasopharyngeal Neoplasms , Podosomes , Humans , Nasopharyngeal Carcinoma/pathology , Podosomes/metabolism , Podosomes/pathology , Herpesvirus 4, Human/metabolism , Nasopharyngeal Neoplasms/pathology , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Membrane Proteins/metabolism , Viral Matrix Proteins/metabolism , Tumor Microenvironment
3.
Cancer Lett ; 524: 70-81, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34637843

ABSTRACT

Aberrated PLK4 expression has been reported in different malignancies and causes centrosome amplification, aneuploidy, and genomic instability. However, the mechanism by which PLK4 is regulated in carcinogenesis remains not fully characterised. Here, we showed that PLK4 was overexpressed in human HCC and overexpression of PLK4 predicted poorer patient prognosis. Unexpectedly, we found that induced expression of PLK4 promotes, but knockdown of PLK4 inhibits, HCC cell migration and invasion. Mechanistically, we found that TEC tyrosine kinase, which also promotes HCC cell migration, stabilizes PLK4 by phosphorylation. TEC directly phosphorylates PLK4 at tyrosine 86 residue, which not only stabilizes the protein but also enhances PLK4-mediated HCC cell invasion. Further investigation by transcriptome sequencing indicated that PLK4 promotes the phosphorylation of focal adhesion kinase to regulate the focal adhesion pathway in HCC cell migration. Taken together, our results demonstrated that PLK4 plays an important role in HCC metastasis and revealed for the first time the mechanism by which PLK4 promotes HCC metastasis via TEC phosphorylation.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Carcinogenesis/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Gene Expression Regulation, Neoplastic/genetics , Genomic Instability/genetics , Humans , Liver Neoplasms/pathology , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis , Phosphorylation/genetics , Transcriptome/genetics
4.
Cancer Lett ; 492: 147-161, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32827601

ABSTRACT

Liver cancer (hepatocellular carcinoma, HCC) is one of the most prevalent cancers worldwide. Several etiological factors of HCC, including hepatitis B or hepatitis C virus infection, liver cirrhosis and aflatoxin B1 intake has been identified. HBx, which is an oncogenic protein encoded by the hepatitis B virus, is strongly associated with hepatocarcinogenesis. Using stable HBx-expressing cell, we showed that HBx induced chromosome gain, with amplification of centrosomes numbers and deregulation of centrosome ultrastructure. To dissect the mechanism for chromosome instability, our result revealed that HBx contributed to a hyperactive centrosome-microtubule dynamics by accelerating microtubule nucleation and polymerization. Further investigations suggested that HBx interacted with a centrosome linker protein TAX1BP2, which has previously been shown to function as an intrinsic block of centrosome amplification and a tumour suppressor in HCC. Restoring TAX1BP2 was able to block HBx-mediated centrosome amplification and abolish the HBx-mediated centrosome aberration, thereby suppressing chromosome instability. Thus, we demonstrate here a mechanism by which HBx deregulates centrosome-microtubule dynamics through interacting with TAX1BP2, which underlines the possibility of restoration of TAX1BP2 to rescue cells from chromosome instability.


Subject(s)
Carcinoma, Hepatocellular/etiology , Centrosome/physiology , Chromosomal Instability , Intracellular Signaling Peptides and Proteins/physiology , Liver Neoplasms/etiology , Membrane Proteins/physiology , Microtubules/physiology , Trans-Activators/physiology , Tumor Suppressor Proteins/physiology , Viral Regulatory and Accessory Proteins/physiology , Adult , Aneuploidy , Hep G2 Cells , Humans , Male
5.
Cell Rep ; 25(3): 690-701.e8, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30332648

ABSTRACT

Arginine methylation is a post-translational modification that plays pivotal roles in signal transduction and gene transcription during cell fate determination. We found protein methyltransferase 6 (PRMT6) to be frequently downregulated in hepatocellular carcinoma (HCC) and its expression to negatively correlate with aggressive cancer features in HCC patients. Silencing of PRMT6 promoted the tumor-initiating, metastasis, and therapy resistance potential of HCC cell lines and patient-derived organoids. Consistently, loss of PRMT6 expression aggravated liver tumorigenesis in a chemical-induced HCC PRMT6 knockout (PRMT6-/-) mouse model. Integrated transcriptome and protein-protein interaction studies revealed an enrichment of genes implicated in RAS signaling and showed that PRMT6 interacted with CRAF on arginine 100, which decreased its RAS binding potential and altered its downstream MEK/ERK signaling. Our work describes a critical repressive function for PRMT6 in maintenance of HCC cells by regulating RAS binding and MEK/ERK signaling via methylation of CRAF on arginine 100.


Subject(s)
Carcinoma, Hepatocellular/pathology , DNA Methylation , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/pathology , Nuclear Proteins/metabolism , Protein-Arginine N-Methyltransferases/metabolism , TNF Receptor-Associated Factor 3/genetics , Animals , Apoptosis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Knockout , Mice, Nude , Mice, SCID , Neoplastic Stem Cells/metabolism , Nuclear Proteins/genetics , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/physiology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , raf Kinases/genetics , raf Kinases/metabolism , ras Proteins/genetics , ras Proteins/metabolism
6.
Proteomics Clin Appl ; 11(3-4)2017 03.
Article in English | MEDLINE | ID: mdl-27801555

ABSTRACT

PURPOSE: Our previous results showed that cadmium (Cd)-adapted lung epithelial cells (LECs) developed resistance to apoptosis due to non-responsiveness of the c-Jun N-terminal kinase pathway and augmented expression of cytokeratin 8. Since cellular Cd entry is a prerequisite in order for Cd to elicit its cytotoxicity, therefore, we wonder if there are differential metal ion transport ability and also other phenotypic changes that occurred in these Cd-resistant LECs. EXPERIMENTAL DESIGN AND RESULTS: Here, we explored further and found that the zinc (Zn) importer Zip8 was stably abolished in these cells along with a marked decrease of Cd and Zn accumulation. Moreover, by cell migration assays and cytokine antibody array analysis, we found that Cd-adapted cells exhibit enhanced migratory ability possibly due to elevated secretions of vascular endothelial growth factor and macrophage inflammatory protein-3 alpha (MIP-3α). CONCLUSION AND CLINICAL RELEVANCE: Taken together, our results show that during chronic Cd exposure, lung cells antagonize excessive cellular Cd-influx by abolishing Zip8 expression to reduce Cd-toxicity; however, this also renders cells with a diminished Zn uptake. The imbalance of Zn homeostasis and elevation of angiogenic and epithelial-mesenchymal transition-promoting cytokines in Cd-adapted cells might thus likely promote Zn deficiency, angiogenesis, and cell invasion.


Subject(s)
Cadmium/toxicity , Cytokines/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Lung/cytology , Zinc/metabolism , Adaptation, Physiological/drug effects , Antibodies, Neutralizing/immunology , Biological Transport/drug effects , Biomarkers/metabolism , Cation Transport Proteins/metabolism , Cell Movement/drug effects , Chemokine CCL20/immunology , Epithelial Cells/cytology , Humans , Time Factors , Vascular Endothelial Growth Factor A/immunology
7.
Cancer Res ; 76(19): 5732-5742, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27496712

ABSTRACT

PAK4 kinase contributes to signaling pathways controlling cancer cell transformation, invasion, and survival, but its clinicopathological impact has begun to emerge only recently. Here we report that PAK4 overexpression in hepatocellular carcinoma (HCC) conveys aggressive metastatic properties. A novel nuclear splice isoform of PAK4 lacking exon 2 sequences was isolated as part of our studies. By stably overexpressing or silencing PAK4 in HCC cells, we showed that it was critical for their migration. Mechanistic investigations in this setting revealed that PAK4 directly phosphorylated p53 at S215, which not only attenuated transcriptional transactivation activity but also inhibited p53-mediated suppression of HCC cell invasion. Taken together, our results showed how PAK4 overexpression in HCC promotes metastatic invasion by regulating p53 phosphorylation. Cancer Res; 76(19); 5732-42. ©2016 AACR.


Subject(s)
Liver Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , p21-Activated Kinases/physiology , Cell Line, Tumor , Cell Movement , DNA/metabolism , Humans , Neoplasm Metastasis , Phosphorylation , Serine/metabolism , p21-Activated Kinases/analysis
8.
Oncotarget ; 7(17): 24005-17, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27006468

ABSTRACT

Tumor relapse after chemotherapy typifies hepatocellular carcinoma (HCC) and is believed to be attributable to residual cancer stem cells (CSCs) that survive initial treatment. Chronic infection with hepatitis B virus (HBV) has long been linked to the development of HCC. Upon infection, random HBV genome integration can lead to truncation of hepatitis B virus X (HBx) protein at the C-terminus. The resulting C-terminal-truncated HBx (HBx-ΔC) was previously shown to confer enhanced invasiveness and diminished apoptotic response in HCC cells. Here, we found HBx-ΔC to promote the appearance of a CD133 liver CSC subset and confer cancer and stem cell-like features in HCC. HBx-ΔC was exclusively detected in HCC cell lines that were raised from patients presented with a HBV background with concomitant CD133 expression. Stable overexpression of the naturally occurring HBx-ΔC mutants, HBx-Δ14 or HBx-Δ35, in HCC cells Huh7 and immortalized normal liver cells MIHA resulted in a significant increase in the cells ability to self-renew, resist chemotherapy and targeted therapy, migrate and induce angiogenesis. MIHA cells with the mutants stably overexpressed also resulted in the induction of CD133, mediated through STAT3 activation. RNA sequencing profiling of MIHA cells with or without HBx-ΔC mutants stably overexpressed identified altered FXR activation. This, together with rescue experiments using a selective FXR inhibitor suggested that C-terminal truncated HBx can mediate cancer stemness via FXR activation. Collectively, we find C-terminal truncated HBx mutants to confer cancer and stem cell-like features in vitro and to play an important role in driving tumor relapse in HCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinogenesis/pathology , Carcinoma, Hepatocellular/pathology , Hepatitis B/pathology , Liver Neoplasms/pathology , Neoplastic Stem Cells/pathology , Trans-Activators/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Cycle , Cell Proliferation , Genome, Viral , Hepatitis B/genetics , Hepatitis B/metabolism , Hepatitis B virus/physiology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Protein Domains , Trans-Activators/genetics , Tumor Cells, Cultured , Viral Regulatory and Accessory Proteins
9.
Oncotarget ; 7(5): 6146-58, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26716417

ABSTRACT

Protein array technology is a powerful platform for the simultaneous determination of the expression levels of a number of proteins as well as post-translational modifications such as phosphorylation. Here, we screen and report for the first time, the dominant signaling cascades and apoptotic mediators during the course of cadmium (Cd)-induced cytotoxicity in human bronchial epithelial cells (BEAS-2B) by antibody array analyses. Proteins from control and Cd-treated cells were captured on Proteome Profiler™ Arrays for the parallel determination of the relative levels of protein phosphorylation and proteins associated with apoptosis. Our results indicated that the p38 MAPK- and JNK-related signal transduction pathways were dramatically activated by Cd treatment. Cd potently stimulates the phosphorylations of p38α (MAPK14), JNK1/2 (MAPK8/9), and JUN; while the phosphorylations of Akt1, ERK1/2 (MAPK3/1), GSK3ß, and mTOR were suppressed. Moreover, there was an induction of proapoptotic protein BAX, release of cytochrome c (CYCS) from mitochondria, activation of caspase-3/9 (CASP3/9); as well as decreased expression of cell cycle checkpoint proteins (TP53, p21, and p27) and several inhibitors of apoptosis proteins (IAPs) [including cIAP-1/2 (BIRC2/3), XIAP (BIRC4), and survivin (BIRC5)]. Pretreatment of cells with the thiol antioxidant glutathione or p38 MAPK/JNK inhibitors before Cd treatment effectively abrogated ROS activation of p38 MAPK/JNK pathways and apoptosis-related proteins. Taken together, our results demonstrate that Cd causes oxidative stress-induced apoptosis; and the p38 MAPK/JNK and mitochondrial pathways are more importantly participated for signal transduction and the induction of apoptosis in Cd-exposed human lung cells.


Subject(s)
Bronchi/drug effects , Bronchi/metabolism , Cadmium Poisoning/metabolism , Proteome/metabolism , Antibodies/chemistry , Apoptosis/drug effects , Cadmium/pharmacology , Cadmium Poisoning/pathology , Cell Line, Tumor , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Phosphorylation , Protein Array Analysis/methods , Protein Processing, Post-Translational , Signal Transduction
10.
Proc Natl Acad Sci U S A ; 111(44): 15810-5, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25331877

ABSTRACT

Adiponectin (ADN) is an adipocyte-secreted protein with insulin-sensitizing, antidiabetic, antiinflammatory, and antiatherogenic properties. Evidence is also accumulating that ADN has neuroprotective activities, yet the underlying mechanism remains elusive. Here we show that ADN could pass through the blood-brain barrier, and elevating its levels in the brain increased cell proliferation and decreased depression-like behaviors. ADN deficiency did not reduce the basal hippocampal neurogenesis or neuronal differentiation but diminished the effectiveness of exercise in increasing hippocampal neurogenesis. Furthermore, exercise-induced reduction in depression-like behaviors was abrogated in ADN-deficient mice, and this impairment in ADN-deficient mice was accompanied by defective running-induced phosphorylation of AMP-activated protein kinase (AMPK) in the hippocampal tissue. In vitro analyses indicated that ADN itself could increase cell proliferation of both hippocampal progenitor cells and Neuro2a neuroblastoma cells. The neurogenic effects of ADN were mediated by the ADN receptor 1 (ADNR1), because siRNA targeting ADNR1, but not ADNR2, inhibited the capacity of ADN to enhance cell proliferation. These data suggest that adiponectin may play a significant role in mediating the effects of exercise on hippocampal neurogenesis and depression, possibly by activation of the ADNR1/AMPK signaling pathways, and also raise the possibility that adiponectin and its agonists may represent a promising therapeutic treatment for depression.


Subject(s)
Adipocytes/metabolism , Adiponectin/metabolism , Depression/metabolism , Hippocampus/metabolism , Neurogenesis , Physical Conditioning, Animal , AMP-Activated Protein Kinases/metabolism , Adipocytes/pathology , Adiponectin/agonists , Animals , Antidepressive Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation , Depression/drug therapy , Hippocampus/pathology , Mice , Phosphorylation , Receptors, Adiponectin/metabolism , Signal Transduction
11.
J Mol Signal ; 9: 7, 2014.
Article in English | MEDLINE | ID: mdl-25093037

ABSTRACT

The p21-activated kinases (PAKs) are downstream effectors of the Rho family small GTPases as well as a wide variety of mitogenic factors and have been implicated in cancer formation, development and metastasis. PAKs phosphorylate a wide spectrum of substrates to mediate extracellular signals and regulate cytoskeletal remodeling, cell motility and survival. In this review, we aim to summarize the findings regarding the oncogenic role and the underlying mechanisms of PAKs signaling in various cancers, and in particular highlight the prime importance of PAKs in hepatocellular carcinoma (HCC) progression and metastasis. Recent studies exploring the potential therapeutic application of PAK inhibitors will also be discussed.

12.
Sci Rep ; 4: 5587, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24998389

ABSTRACT

This study aimed to investigate the possible therapeutic effects and active components of Lycium barbarum polysaccharides (LBP) on a high fat diet-induced NASH rat model. We induced NASH in a rat model by voluntary oral feeding with a high-fat diet ad libitum for 8 weeks. After 8 weeks, 1 mg/kg LBP was orally administered for another 4 weeks with a high-fat diet. When compared with NASH rats treated for 12 weeks, therapeutic LBP treatment for 4 weeks during 12 weeks of NASH induction showed ameliorative effects on: (1) increased body and wet liver weights; (2) insulin resistance and glucose metabolic dysfunction; (3) elevated level of serum aminotransferases; (4) fat accumulation in the liver and increased serum free fatty acid (FFA) level; (5) hepatic fibrosis; (6) hepatic oxidative stress; (7) hepatic inflammatory response; and (8) hepatic apoptosis. These improvements were partially through the modulation of transcription factor NF-κB, MAPK pathways and the autophagic process. In a palmitate acid-induced rat hepatocyte steatosis cell-based model, we also demonstrated that l-arabinose and ß-carotene partially accounted for the beneficial effects of LBP on the hepatocytes. In conclusion, LBP possesses a variety of hepato-protective properties which make it a potent supplementary therapeutic agent against NASH in future clinical trials.


Subject(s)
Anti-Obesity Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Hepatocytes/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Apoptosis , Arabinose/pharmacology , Autophagy , Cell Survival , Cells, Cultured , Diet, High-Fat/adverse effects , Female , Insulin Resistance , Lipid Metabolism/drug effects , Liver/drug effects , Liver/pathology , MAP Kinase Signaling System , Non-alcoholic Fatty Liver Disease/etiology , Obesity/drug therapy , Oxidative Stress , Rats, Sprague-Dawley , beta Carotene/pharmacology
13.
Nat Genet ; 46(6): 573-82, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24816253

ABSTRACT

Gastric cancer is a heterogeneous disease with diverse molecular and histological subtypes. We performed whole-genome sequencing in 100 tumor-normal pairs, along with DNA copy number, gene expression and methylation profiling, for integrative genomic analysis. We found subtype-specific genetic and epigenetic perturbations and unique mutational signatures. We identified previously known (TP53, ARID1A and CDH1) and new (MUC6, CTNNA2, GLI3, RNF43 and others) significantly mutated driver genes. Specifically, we found RHOA mutations in 14.3% of diffuse-type tumors but not in intestinal-type tumors (P < 0.001). The mutations clustered in recurrent hotspots affecting functional domains and caused defective RHOA signaling, promoting escape from anoikis in organoid cultures. The top perturbed pathways in gastric cancer included adherens junction and focal adhesion, in which RHOA and other mutated genes we identified participate as key players. These findings illustrate a multidimensional and comprehensive genomic landscape that highlights the molecular complexity of gastric cancer and provides a road map to facilitate genome-guided personalized therapy.


Subject(s)
Gene Expression Regulation, Neoplastic , Mutation , Stomach Neoplasms/genetics , Adherens Junctions , Algorithms , Animals , DNA Methylation , DNA Mutational Analysis , Epigenesis, Genetic , Female , Gene Dosage , Gene Expression Profiling , Genetic Variation , Genome, Human , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , rhoA GTP-Binding Protein/genetics
14.
PLoS One ; 8(7): e68843, 2013.
Article in English | MEDLINE | ID: mdl-23894351

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the major malignancies worldwide and is associated with poor prognosis due to the high incidences of metastasis and tumor recurrence. Our previous study showed that overexpression of p21-activated protein kinase 1 (PAK1) is frequently observed in HCC and is associated with a more aggressive tumor behavior, suggesting that PAK1 is a potential therapeutic target in HCC. In the current study, an allosteric small molecule PAK1 inhibitor, IPA-3, was evaluated for the potential in suppressing hepatocarcinogenesis. Consistent with other reports, inhibition of PAK1 activity was observed in several human HCC cell lines treated with various dosages of IPA-3. Using cell proliferation, colony formation and BrdU incorporation assays, we demonstrated that IPA-3 treatment significantly inhibited the growth of HCC cells. The mechanisms through which IPA-3 treatment suppresses HCC cell growth are enhancement of apoptosis and blockage of activation of NF-κB. Furthermore, our data suggested that IPA-3 not only inhibits the HCC cell growth, but also suppresses the metastatic potential of HCC cells. Nude mouse xenograft assay demonstrated that IPA-3 treatment significantly reduced the tumor growth rate and decreased tumor volume, indicating that IPA-3 can suppress the in vivo tumor growth of HCC cells. Taken together, our demonstration of the potential preclinical efficacy of IPA-3 in HCC provides the rationale for cancer therapy.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Disulfides/pharmacology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , NF-kappa B/metabolism , Naphthols/pharmacology , p21-Activated Kinases/metabolism , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Enzyme Activation/drug effects , Humans , Male , Protein Transport , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
Article in English | MEDLINE | ID: mdl-23861709

ABSTRACT

Our previous study demonstrated that administration of garlic-derived antioxidant S-allylmercaptocysteine (SAMC) ameliorated hepatic injury in a nonalcoholic fatty liver disease (NAFLD) rat model. Our present study aimed to investigate the mechanism of SAMC on NAFLD-induced hepatic apoptosis and autophagy. Adult female rats were fed with a high-fat diet for 8 weeks to develop NAFLD with or without intraperitoneal injection of 200 mg/kg SAMC for three times per week. During NAFLD development, increased apoptotic cells and caspase-3 activation were observed in the liver. Increased apoptosis was modulated through both intrinsic and extrinsic apoptotic pathways. NAFLD treatment also enhanced the expression of key autophagic markers in the liver with reduced activity of LKB1/AMPK and PI3K/Akt pathways. Increased expression of proapoptotic regulator p53 and decreased activity of antiautophagic regulator mTOR were also observed. Administration of SAMC reduced the number of apoptotic cells through downregulation of both intrinsic and extrinsic apoptotic mechanisms. SAMC also counteracted the effects of NAFLD on LKB1/AMPK and PI3K/Akt pathways. Treatment with SAMC further enhanced hepatic autophagy by regulating autophagic markers and mTOR activity. In conclusion, administration of SAMC during NAFLD development in rats protects the liver from chronic injury by reducing apoptosis and enhancing autophagy.

16.
Retrovirology ; 10: 47, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23622267

ABSTRACT

BACKGROUND: Human T-cell leukemia virus type 1 (HTLV-1) is the causative agent of adult T-cell leukemia and tropical spastic paraparesis. HTLV-1 encodes transactivator protein Tax that interacts with various cellular factors to modulate transcription and other biological functions. Additional cellular mediators of Tax-mediated transcriptional activation of HTLV-1 long terminal repeats (LTR) remain to be identified and characterized. RESULTS: In this study, we investigated the regulatory role of group I p21-activated kinases (Paks) in Tax-induced LTR activation. Both wild-type and kinase-dead mutants of Pak3 were capable of potentiating the activity of Tax to activate LTR transcription. The effect of Paks on the LTR was attributed to the N-terminal regulatory domain and required the action of CREB, CREB-regulating transcriptional coactivators (CRTCs) and p300/CREB-binding protein. Paks physically associated with Tax and CRTCs. Paks were recruited to the LTR in the presence of Tax. siRNAs against either Pak1 or Pak3 prevented the interaction of Tax with CRTC1 and the recruitment of Tax to the LTR. These siRNAs also inhibited LTR-dependent transcription in HTLV-1-transformed MT4 cells and in cells transfected with an infectious clone of HTLV-1. CONCLUSION: Group I Paks augment Tax-mediated transcriptional activation of HTLV-1 LTR in a kinase-independent manner.


Subject(s)
Gene Products, tax/metabolism , Host-Pathogen Interactions , Human T-lymphotropic virus 1/physiology , Terminal Repeat Sequences , Transcriptional Activation , Virus Replication , p21-Activated Kinases/metabolism , HeLa Cells , Humans , Protein Binding , Protein Interaction Mapping
17.
Retrovirology ; 10: 40, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23577667

ABSTRACT

BACKGROUND: Human T-cell leukemia virus type 1 (HTLV-1) causes adult T-cell leukemia (ATL). Treatment options are limited and prophylactic agents are not available. We have previously demonstrated an essential role for CREB-regulating transcriptional coactivators (CRTCs) in HTLV-1 transcription. RESULTS: In this study we report on the negative regulatory role of LKB1 tumor suppressor and salt-inducible kinases (SIKs) in the activation of HTLV-1 long terminal repeats (LTR) by the oncoprotein Tax. Activation of LKB1 and SIKs effectively blunted Tax activity in a phosphorylation-dependent manner, whereas compromising these kinases, but not AMP-dependent protein kinases, augmented Tax function. Activated LKB1 and SIKs associated with Tax and suppressed Tax-induced LTR activation by counteracting CRTCs and CREB. Enforced expression of LKB1 or SIK1 in cells transfected with HTLV-1 molecular clone pX1MT repressed proviral transcription. On the contrary, depletion of LKB1 in pX1MT-transfected cells and in HTLV-1-transformed T cells boosted the expression of Tax. Treatment of HTLV-1 transformed cells with metformin led to LKB1/SIK1 activation, reduction in Tax expression, and inhibition of cell proliferation. CONCLUSIONS: Our findings revealed a new function of LKB1 and SIKs as negative regulators of HTLV-1 transcription. Pharmaceutical activation of LKB1 and SIKs might be considered as a new strategy in anti-HTLV-1 and anti-ATL therapy.


Subject(s)
Gene Expression Regulation, Viral , Gene Products, tax/antagonists & inhibitors , Host-Pathogen Interactions , Human T-lymphotropic virus 1/physiology , Protein Serine-Threonine Kinases/metabolism , Transcription, Genetic , AMP-Activated Protein Kinase Kinases , Cell Line , Humans
18.
Eur J Nutr ; 52(1): 179-91, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22278044

ABSTRACT

PURPOSE: To investigate the hepato-protective properties and underlying mechanisms of SAMC in a non-alcoholic fatty liver disease (NAFLD) rat model. METHODS: Female rats were fed with a diet comprising highly unsaturated fat diet (30% fish oil) for 8 weeks to develop NAFLD with or without an intraperitoneal injection of 200 mg/kg SAMC three times per week. After euthanasia, blood and liver samples of rats were collected for histological and biochemical analyses. RESULTS: Co-treatment of SAMC attenuated NAFLD-induced liver injury, fat accumulation, collagen formation and free fatty acids (FFAs). At the molecular level, SAMC decreased the lipogenesis marker and restored the lipolysis marker. SAMC also reduced the expression levels of pro-fibrogenic factors and diminished liver oxidative stress partly through the inhibition in the activity of cytochrome P450 2E1-dependent pathway. NAFLD-induced inflammation was also partially mitigated by SAMC treatment via reduction in the pro-inflammatory mediators, chemokines and suppressor of cytokine signaling. The protective effect of SAMC is also shown partly through the restoration of altered phosphorylation status of FFAs-dependent MAP kinase pathways and diminished in the nuclear transcription factors (NF-κB and AP-1) activity during NAFLD development. CONCLUSIONS: SAMC is a novel hepato-protective agent against NAFLD caused by abnormal liver functions. Garlic or garlic derivatives could be considered as a potent food supplement in the prevention of fatty liver disease.


Subject(s)
Cysteine/analogs & derivatives , Fatty Liver/drug therapy , Garlic/chemistry , Liver/drug effects , Plant Extracts/pharmacology , Animals , Blotting, Western , Cysteine/pharmacology , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , Cytochrome P-450 CYP2E1 Inhibitors , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Inflammation/drug therapy , Inflammation/pathology , Lipogenesis/drug effects , Liver/metabolism , MAP Kinase Signaling System/drug effects , NF-kappa B/genetics , NF-kappa B/metabolism , Non-alcoholic Fatty Liver Disease , Oxidative Stress/drug effects , Phosphorylation , Rats , Rats, Sprague-Dawley , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
19.
PLoS One ; 7(7): e42210, 2012.
Article in English | MEDLINE | ID: mdl-22860085

ABSTRACT

CDK5 regulatory subunit associated protein 3 (CDK5RAP3) is a novel activator of PAK4 and processes important pro-metastatic function in hepatocarcinogenesis. However, it remains unclear if there are other mechanisms by which CDK5RAP3 promotes HCC metastasis. Here, we showed that in CDK5RAP3 stable knockdown SMMC-7721 HCC cells, p14(ARF) tumor suppressor was upregulated at protein and mRNA levels, and ectopic expression of CDK5RAP3 was found to repress the transcription of p14(ARF). Using chromatin immunoprecipitation assay, we demonstrated that CDK5RAP3 bound to p14(ARF) promoter in vivo. Furthermore, knockdown of p14(ARF) in CDK5RAP3 stable knockdown HCC cells reversed the suppression of HCC cell invasiveness mediated by knockdown of CDK5RAP3. Taken together, our findings provide the new evidence that overexpression of CDK5RAP3 promotes HCC metastasis via downregulation of p14(ARF).


Subject(s)
Carcinoma, Hepatocellular/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Liver Neoplasms/metabolism , Nerve Tissue Proteins/physiology , Tumor Suppressor Protein p14ARF/physiology , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins , Cell Line, Tumor , Chromatin Immunoprecipitation , Gene Knockdown Techniques , Humans , Liver Neoplasms/pathology , Microscopy, Confocal , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Proteins
20.
Cancer Res ; 72(17): 4394-404, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22728651

ABSTRACT

AMP-activated protein kinase (AMPK), a biologic sensor for cellular energy status, has been shown to act upstream and downstream of known tumor suppressors. However, whether AMPK itself plays a tumor suppressor role in cancer remains unclear. Here, we found that the α2 catalytic subunit isoform of AMPK is significantly downregulated in hepatocellular carcinoma (HCC). Clinicopathologic analysis revealed that underexpression of AMPK-α2 was statistically associated with an undifferentiated cellular phenotype and poor patient prognosis. Loss of AMPK-α2 in HCC cells rendered them more tumorigenic than control cells both in vitro and in vivo. Mechanistically, ectopic expression of AMPK enhanced the acetylation and stability of p53 in HCC cells. The p53 deacetylase, SIRT1, was phosphorylated and inactivated by AMPK at Thr344, promoting p53 acetylation and apoptosis of HCC cells. Taken together, our findings suggest that underexpression of AMPK is frequently observed in HCC, and that inactivation of AMPK promotes hepatocarcinogenesis by destabilizing p53 in a SIRT1-dependent manner.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Sirtuin 1/genetics , Tumor Suppressor Protein p53/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/genetics , Acetylation , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/pharmacology , Sirtuin 1/metabolism , Transcriptional Activation , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...