Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 75(17): 3568-82, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26100670

ABSTRACT

Mitochondria are the powerhouses of cells. Mitochondrial C-Raf is a potential cancer therapeutic target, as it regulates mitochondrial function and is localized to the mitochondria by its N-terminal domain. However, Raf inhibitor monotherapy can induce S338 phosphorylation of C-Raf (pC-Raf(S338)) and impede therapy. This study identified the interaction of C-Raf with S308 phosphorylated DAPK (pDAPK(S308)), which together became colocalized in the mitochondria to facilitate mitochondrial remodeling. Combined use of the Raf inhibitors sorafenib and GW5074 had synergistic anticancer effects in vitro and in vivo, but targeted mitochondrial function, rather than the canonical Raf signaling pathway. C-Raf depletion in knockout MEF(C-Raf-/-) or siRNA knockdown ACHN renal cancer cells abrogated the cytotoxicity of combination therapy. Crystal structure simulation showed that GW5074 bound to C-Raf and induced a C-Raf conformational change that enhanced sorafenib-binding affinity. In the presence of pDAPK(S308), this drug-target interaction compromised the mitochondrial targeting effect of the N-terminal domain of C-Raf, which induced two-hit damages to cancer cells. First, combination therapy facilitated pC-Raf(S338) and pDAPK(S308) translocation from mitochondria to cytoplasm, leading to mitochondrial dysfunction and reactive oxygen species (ROS) generation. Second, ROS facilitated PP2A-mediated dephosphorylation of pDAPK(S308) to DAPK. PP2A then dissociated from the C-Raf-DAPK complex and induced profound cancer cell death. Increased pDAPK(S308) modification was also observed in renal cancer tissues, which correlated with poor disease-free survival and poor overall survival in renal cancer patients. Besides mediating the anticancer effect, pDAPK(S308) may serve as a predictive biomarker for Raf inhibitors combination therapy, suggesting an ideal preclinical model that is worthy of clinical translation.


Subject(s)
Death-Associated Protein Kinases/genetics , Drug Synergism , Kidney Neoplasms/drug therapy , Proto-Oncogene Proteins c-raf/genetics , Aged , Animals , Apoptosis/drug effects , Cell Line, Tumor , Disease-Free Survival , Female , Gene Knockout Techniques , Humans , Indoles/administration & dosage , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Mice , Middle Aged , Mitochondria/drug effects , Mitochondria/pathology , Niacinamide/administration & dosage , Niacinamide/analogs & derivatives , Phenols/administration & dosage , Phenylurea Compounds/administration & dosage , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sorafenib , Xenograft Model Antitumor Assays
2.
Mol Carcinog ; 54(3): 167-77, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24115089

ABSTRACT

The deregulation of epigenetics was involved in early and subsequent carcinogenic events. Reversing cancer epigenetics to restore a normal epigenetic condition could be a rational approach for cancer treatment and specialized prevention. In the present study, we found that the expression levels of two epigenetic markers, histone H3K27 trimethylation (H3K27me3), was low but histone H3S10 phosphorylation (pH3Ser10) was high in human bladder cancer tissues, which showed opposite expression patterns in their normal counterparts. Thus, we investigated whether a natural product, emodin, has the ability to reverse these two epigenetic modifications and inhibit bladder cancer cell growth. Emodin significantly inhibited the cell growth of four bladder cancer cell lines in a dose- and time-dependent manner. Emodin treatment did not induce specific cell cycle arrest, but it altered epigenetic modifications. Emodin treatment resulted in the suppression of pH3Ser10 and increased H3K27me3, contributing to gene silencing in bladder cancer cells. Microarray analysis demonstrated that oncogenic genes including fatty acid binding protein 4 (FABP4) and fibroblast growth factor binding protein 1 (HBP17), RGS4, tissue inhibitor of metalloproteinase 3 (TIMP3), WNT5b, URB, and collagen, type VIII, alpha 1 (COL8A1) responsible for proliferation, survival, inflammation, and carcinogenesis were significantly repressed by emodin. The ChIP assays also showed that emodin increased H3K27me3 but decreased pH3Ser10 modifications on the promoters of repressed genes, which indicate that emodin reverses the cancer epigenetics towards normal epigenetic situations. In conclusion, our work demonstrates the significant anti-neoplastic activity of emodin on bladder cancer cells and elucidates the novel mechanisms of emodin-mediated epigenetic modulation of target genes. Our study warrants further investigation of emodin as an effective therapeutic or preventive agent for bladder cancer.


Subject(s)
Emodin/pharmacology , Epigenesis, Genetic/drug effects , Protein Kinase Inhibitors/pharmacology , Urinary Bladder Neoplasms/drug therapy , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , DNA Methylation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Histones/genetics , Histones/metabolism , Humans , Phosphorylation , Promoter Regions, Genetic/drug effects , Tumor Cells, Cultured , Urothelium/cytology , Urothelium/metabolism
3.
Oncotarget ; 5(21): 10342-55, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25431950

ABSTRACT

The polycomb group gene, EZH2, is highly expressed in advanced bladder cancer. Here we demonstrated that down-regulation of EZH2 in tumor tissues after neo-adjuvant chemotherapy correlated with good therapeutic response in advanced bladder cancer. We next developed a small molecule, NSC745885, derived from natural anthraquinone emodin, which down-regulated EZH2 via proteasome-mediated degradation. NSC745885 showed potent selective toxicity against multiple cancer cell lines but not normal cells. NSC745885 treatment overcame multiple-drug resistance and inhibited growth of resistant cancer cells. Over-expression of EZH2 in cancer cells attenuated effects of NSC745885, suggesting that down-regulation of EZH2 was responsible for growth inhibition of NSC745885. NSC745885 also suppressed tumor growth and down-regulated EZH2 in vivo. These results indicate that NSC7455889 suppresses bladder cancer by targeting EZH2.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma/drug therapy , Emodin/administration & dosage , Neoadjuvant Therapy , Polycomb Repressive Complex 2/metabolism , Proteasome Endopeptidase Complex/metabolism , Urinary Bladder Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Down-Regulation , Drug Resistance/drug effects , Emodin/analogs & derivatives , Emodin/pharmacology , Enhancer of Zeste Homolog 2 Protein , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , In Vitro Techniques , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Staging , Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/genetics , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
4.
PLoS One ; 8(9): e73401, 2013.
Article in English | MEDLINE | ID: mdl-24023871

ABSTRACT

Histone deacetylase inhibitors (HDACIs) have potent anti-cancer activity in a variety of cancer models. Understanding the molecular mechanisms involved in the therapeutic responsiveness of HDACI is needed before its clinical application. This study aimed to determine if a potent HDACI, LBH589 (Panobinostat), had differential therapeutic responsiveness towards LNCaP and PC-3 prostate cancer (PCa) cells. The former showed prometaphase arrest with subsequent apoptosis upon LBH589 treatment, while the latter was less sensitive and had late G2 arrest. The LBH589 treatment down-regulated HDAC6 and sustained ERK activation, and contributed to prometaphase arrest. Mechanistically, LBH589 inhibited HDAC6 activity, caused its dissociation from protein phosphatase PP1α, and increased 14-3-3ζ acetylation. Acetylated 14-3-3ζ released its mask effect on serine 259 of c-Raf and serine 216 of Cdc25C subsequent to de-phosphorylation by PP1α, which contributed to ERK activation. Enhanced ERK activity by LBH589 further down-regulated HDAC6 protein levels and sustained ERK activation by free-forward regulation. The sustained Cdc25C and ERK activation resulted in early M-phase (prometaphase) arrest and subsequent apoptosis in the most sensitive LNCaP cells but not in PC-3 cells. This study provides pre-clinical evidence that HDAC6 may serve as a sensitive therapeutic target in the treatment of prostate cancer with HDACI LBH589 for clinical translation. This study also posits a novel mechanism of HDAC6 participation in regulating the c-Raf-PP1-ERK signaling pathway and contributing to M phase cell-cycle transition.


Subject(s)
Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Indoles/pharmacology , M Phase Cell Cycle Checkpoints/drug effects , Prostatic Neoplasms/pathology , 14-3-3 Proteins/metabolism , Acetylation/drug effects , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Histone Deacetylase 6 , Histone Deacetylase Inhibitors/pharmacology , Humans , Male , Panobinostat , Protein Phosphatase 1/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Signal Transduction/drug effects , cdc25 Phosphatases/metabolism
5.
Mol Cancer Res ; 10(8): 1109-19, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22707636

ABSTRACT

TNF-α is a cytokine with antitumorigenic property. In contrast, low dose, chronic TNF-α production by tumor cells or stromal cells may promote tumor growth and metastasis. Serum levels of TNF-α are significantly elevated in renal cell carcinoma (RCC) patients. Here, we showed that TNF-α induced epithelial-mesenchymal transition (EMT) and promoted tumorigenicity of RCC by repressing E-cadherin, upregulating vimentin, activating MMP9, and invasion activities. In addition, TNF-α treatment inhibited glycogen synthase kinase 3ß (GSK-3ß) activity through serine-9 phosphorylation mediated by the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) pathway in RCC cells. Inhibition of PI3K/AKT by LY294002 reactivated GSK-3ß and suppressed the TNF-α-induced EMT of RCC cells. Inactivation of GSK-3ß by LiCl significantly increased MMP9 activity and EMT of RCC cells. Activation of GSK-3ß by transduction of constitutively active GSK-3ß into RCC cells suppressed TNF-α-mediated anchorage-independent growth in soft agar and tumorigenicity in nude mice. Overexpression of a kinase-deficient GSK-3ß, in contrast, potentiated EMT, anchorage-independent growth and drastically enhanced tumorigenicity in vivo. Most importantly, a 15-fold inactivation of GSK-3ß activity, 3-fold decrease of E-cadherin, and 2-fold increase of vimentin were observed in human RCC tumor tissues. These results indicated that inactivation of GSK-3ß plays a pivotal role in the TNF-α-mediated tumorigenesis of RCC.


Subject(s)
Carcinoma, Renal Cell , Cell Transformation, Neoplastic , Glycogen Synthase Kinase 3/metabolism , Kidney Neoplasms , Tumor Necrosis Factor-alpha , Animals , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Chromones/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Morpholines/pharmacology , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Vimentin/genetics , Vimentin/metabolism
6.
Mol Cell Biol ; 31(5): 912-23, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21173162

ABSTRACT

The synaptonemal complex (SC) is a meiosis-specific tripartite structure that forms between two homologous chromosomes; it consists of a central region and two parallel lateral elements. Lateral elements also are called axial elements prior to synapsis. In Saccharomyces cerevisiae, Red1, Hop1, and Mek1 are structural components of axial/lateral elements. The red1/mek1/hop1 mutants all exhibit reduced levels of interhomolog recombination and produce no viable spores. Red1 is a phosphoprotein. Several earlier reports proposed that phosphorylated Red1 plays important roles in meiosis, including in signaling meiotic DNA damage or in preventing exit from the pachytene chromosomes. We report here that the phosphorylation of Red1 is carried out in CDC28-dependent and CDC28-independent manners. In contrast to previous results, we found Red1 phosphorylation to be independent of meiotic DNA recombination, the Mec1/Tel1 DNA damage checkpoint kinases, and the Mek1 kinase. To functionally validate the phosphorylation of Red1, we mapped the phosphorylation sites on this protein. A red1(14A) mutant showing no detectable Red1 phosphorylation did not exhibit decreased sporulation efficiency, defects in viable spore production, or defects in meiotic DNA damage checkpoints. Thus, our results suggest that the phosphorylation of Red1 is not essential for its functions in meiosis.


Subject(s)
Meiosis/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/physiology , Amino Acid Sequence , CDC28 Protein Kinase, S cerevisiae/metabolism , Chromosome Mapping , Chromosome Pairing/genetics , DNA Damage , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase 1/metabolism , Molecular Sequence Data , Mutation , Phosphorylation/genetics , Protein Serine-Threonine Kinases/metabolism , Recombination, Genetic , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Spores, Fungal/genetics , Spores, Fungal/metabolism
7.
Clin Cancer Res ; 15(3): 840-50, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19188154

ABSTRACT

PURPOSE: This study is aimed at investigating antineoplastic efficacy of histone deacetylase inhibitor (HDACI) LBH589 on renal cell carcinoma (RCC) and elucidating the novel molecular mechanisms involved in growth arrest and apoptosis by targeting the important nonhistone molecules. EXPERIMENTAL DESIGN: We analyzed the growth-inhibitory effect of LBH589 on RCC by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay in vitro and antitumor efficacy by xenograft experiments in vivo. To verify the associated molecular mechanisms involved in LBH589-mediated cell death and cell cycle progression by Western blotting and fluorescence-activated cell sorting analysis. RESULTS: HDACI LBH589 induced degradation of both Aurora A and B kinases through a proteasome-mediated pathway by targeting HDAC3 and HDAC6. The dual degradation of Aurora A and B kinases mediated by LBH589 resulted in inducing G2-M arrest and apoptosis of renal cancer cell lines and our results also showed that LBH589 potently inhibited renal cancer cell growth in vitro and suppressed tumor formation in vivo. The Aurora A and B kinases and HDAC3 are overexpressed in the human RCC tumor tissues examined, which make them perfect targets for HDACI LBH589 treatment. CONCLUSIONS: Our in vitro and in vivo data showed that LBH589 has potent anticancer effect of renal cancer cells. LBH589 and other HDACI treatment resulted in inducing G2-M arrest and apoptosis of renal cancer cells through degradation of Aurora A and B kinases by inhibition of HDAC3 and HDAC6. The clinical efficacy of LBH589 in the treatment of patients with metastatic RCC, especially those with high Aurora kinase and HDAC expression, is worthy of further investigation.


Subject(s)
Carcinoma, Renal Cell/drug therapy , G2 Phase , Hydroxamic Acids/therapeutic use , Kidney Neoplasms/drug therapy , Protein Serine-Threonine Kinases/metabolism , Animals , Aurora Kinase A , Aurora Kinases , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Histone Deacetylase 6 , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Humans , Indoles , Kidney Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Panobinostat , Xenograft Model Antitumor Assays
8.
Cancer Sci ; 99(5): 905-13, 2008 May.
Article in English | MEDLINE | ID: mdl-18294286

ABSTRACT

Pro-inflammatory cytokines and chemokines are involved in promoting tumorigenesis by facilitating tumor proliferation and metastasis. The serum levels of interleukin (IL)-6, IL-1 beta, and tumor necrosis factor-alpha (TNF-alpha) are significantly elevated in patients with renal cell carcinoma (RCC). However, the mechanisms of how these cytokines participate in the progression of RCC remains unknown. In the present study, we investigated the effects of tumor-derived cytokines on invasion and the epithelial-mesenchymal transition (EMT) of RCC cells. We found that expression of IL-1 beta, IL-6, TNF-alpha, hypoxia-inducible factor-alpha (HIF-1 alpha), and matrix metalloproteinase-2 (MMP2) were significantly elevated in high malignancy A498 cells compared to low malignancy 786-O cells. The invasion ability of A498 was three-fold higher than that of 786-O cells. The invasiveness of 786-O cells was markedly enhanced by adding conditioned medium derived from A498 cells. This phenomenon was significantly inhibited by immunodepletion of TNF-alpha followed by MMP2, IL-6, or IL-1 beta from A498 conditioned medium. Synergistic inhibition was also noted after simultaneous immunodepletion of TNF-alpha, IL-1 beta, and IL-6. RCC cell lines with higher malignancy produced more TNF-alpha, which was correlated with their stronger invasive ability. The invasiveness of 786-O cells was significantly promoted by TNF-alpha in a dose-dependent manner. Moreover, TNF-alpha induced the EMT of 786-O cells by repressing E-cadherin, promoting vimentin expression, and activating MMP9 activity. Our findings demonstrate that pro-inflammatory cytokines, especially TNF-alpha, can enhance invasion and the EMT of renal cancer cells, which provides a therapeutic target to prevent and treat advanced RCC.


Subject(s)
Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Tumor Necrosis Factor-alpha/metabolism , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/metabolism , Disease Progression , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Kidney Neoplasms/immunology , Kidney Neoplasms/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mesoderm/pathology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...