Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Acta Neuropathol ; 132(6): 917-930, 2016 12.
Article in English | MEDLINE | ID: mdl-27664011

ABSTRACT

Mutant isocitrate dehydrogenase 1 (IDH1) is common in gliomas, and produces D-2-hydroxyglutarate (D-2-HG). The full effects of IDH1 mutations on glioma biology and tumor microenvironment are unknown. We analyzed a discovery cohort of 169 World Health Organization (WHO) grade II-IV gliomas, followed by a validation cohort of 148 cases, for IDH1 mutations, intratumoral microthrombi, and venous thromboemboli (VTE). 430 gliomas from The Cancer Genome Atlas were analyzed for mRNAs associated with coagulation, and 95 gliomas in a tissue microarray were assessed for tissue factor (TF) protein. In vitro and in vivo assays evaluated platelet aggregation and clotting time in the presence of mutant IDH1 or D-2-HG. VTE occurred in 26-30 % of patients with wild-type IDH1 gliomas, but not in patients with mutant IDH1 gliomas (0 %). IDH1 mutation status was the most powerful predictive marker for VTE, independent of variables such as GBM diagnosis and prolonged hospital stay. Microthrombi were far less common within mutant IDH1 gliomas regardless of WHO grade (85-90 % in wild-type versus 2-6 % in mutant), and were an independent predictor of IDH1 wild-type status. Among all 35 coagulation-associated genes, F3 mRNA, encoding TF, showed the strongest inverse relationship with IDH1 mutations. Mutant IDH1 gliomas had F3 gene promoter hypermethylation, with lower TF protein expression. D-2-HG rapidly inhibited platelet aggregation and blood clotting via a novel calcium-dependent, methylation-independent mechanism. Mutant IDH1 glioma engraftment in mice significantly prolonged bleeding time. Our data suggest that mutant IDH1 has potent antithrombotic activity within gliomas and throughout the peripheral circulation. These findings have implications for the pathologic evaluation of gliomas, the effect of altered isocitrate metabolism on tumor microenvironment, and risk assessment of glioma patients for VTE.


Subject(s)
Brain Neoplasms/complications , Brain Neoplasms/genetics , Glioma/complications , Glioma/genetics , Isocitrate Dehydrogenase/genetics , Mutation/genetics , Thrombosis/etiology , Adult , Aged , Aged, 80 and over , Alcohol Oxidoreductases/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Blood Platelets/drug effects , Blood Platelets/metabolism , Brain Neoplasms/drug therapy , Calcimycin/pharmacology , Calcium Ionophores/pharmacology , Cohort Studies , Female , Glioma/drug therapy , Humans , Male , Mice , Middle Aged , Thrombin/metabolism , Thrombin/pharmacology , Thromboplastin/metabolism , Thrombosis/drug therapy , Thrombosis/pathology
2.
Autophagy ; 9(10): 1566-78, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24113030

ABSTRACT

Autophagy resembles a recycling process in which proteins, organelles, or regions of the cytoplasm are enveloped and degraded. We have found that two of the central autophagy proteins, MAP1LC3 (microtubule-associated protein 1 light chain 3, also described as LC3) and UVRAG (UV radiation resistance associated/UV radiation associated gene), complex with PGRMC1/S2R (progesterone receptor membrane component 1, also known as sigma-2 receptor). PGRMC1 is a cytochrome that is induced in cancer and is essential for tumor formation, invasion, and metastasis. Autophagy contributes to the turnover of long-lived and/or ubiquitinated proteins and the clearance of damaged organelles, and we have shown that PGRMC1 promotes both processes. Inhibition of PGRMC1 by RNAi or small molecule inhibitors causes autophagy substrates to increase and aberrant mitochondria to accumulate. We propose that this disruption of autophagy upon PGRMC1 inhibition increases AMPK activation, elevating the levels of TSC1 (tuberous sclerosis complex) and TSC2 and inactivating MTOR and RPS6KB/p70S6K, causing cleaved MAP1LC3B levels to increase. Thus, PGRMC1 binds to key components of the autophagy machinery and is required for the degradative activity of autophagy.


Subject(s)
Autophagy/physiology , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Receptors, Progesterone/metabolism , Receptors, sigma/metabolism , AMP-Activated Protein Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Humans , Proteolysis , Receptors, Progesterone/drug effects , Receptors, sigma/drug effects , Sequestosome-1 Protein , Tuberous Sclerosis/metabolism
3.
Apoptosis ; 18(6): 766-76, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23456622

ABSTRACT

Previous studies demonstrated that proteasome inhibition sensitizes TRAIL resistant prostate cancer cells to TRAIL-mediated apoptosis via stabilization of the active p18 subunit of caspase-8. The present study investigated the impact of proteasome inhibition on caspase-8 stability, ubiquitination, trafficking, and activation in cancer cells. Using caspase-8 deficient neuroblastoma (NB7) cells for reconstituting non-cleavable mutant forms of caspase-8, we demonstrated that the non-cleavable forms of caspase-8 are capable of inducing apoptosis comparably to wild-type caspase-8, in response to proteasome inhibitor and GST-TRAIL. Moreover in the LNCaP human prostate cancer cells, caspase-8 polyubiquitination occurs after TRAIL stimulation and caspase-8 processing. Subcellular fractionation analysis revealed caspase-8 activity in both cytosol and plasma membrane fractions in both NB7 reconstituted caspase-8 cell lines, as well the LNCaP prostate cancer cells. The present results suggest that caspase-8 stabilization through proteasome inhibition leads to reactivation of the extrinsic pathway of apoptosis and identify E3 ligase mediating caspase-8 polyubiquitination, as a novel molecular target. Inhibition of this E3 ligase in combination with TRAIL towards restoring apoptosis signaling activation may have potential therapeutic significance in resistant tumors.


Subject(s)
Apoptosis/drug effects , Caspase 8/metabolism , Proteasome Endopeptidase Complex/metabolism , Caspase 8/genetics , Cell Line, Tumor , Enzyme Activation , Humans , Male , Prostatic Neoplasms/drug therapy , Proteasome Endopeptidase Complex/drug effects , TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Ubiquitination
4.
Mol Cancer ; 10: 57, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21586138

ABSTRACT

BACKGROUND: A functioning ubiquitin proteasome system (UPS) is essential for a number of diverse cellular processes and maintenance of overall cellular homeostasis. The ability of proteasome inhibitors, such as Velcade, to promote extrinsic apoptotic effects illustrates the importance of the ubiquitin proteasome system in the regulation of death receptor signaling. Here, we set out to define the UPS machinery, particularly the E3 ubiquitin ligases, that repress apoptosis through the extrinsic pathway. A cell-based genome-wide E3 ligase siRNA screen was established to monitor caspase-8 activity following the addition of TRAIL. RESULTS: Data from the high-throughput screen revealed that targeting the RING-finger containing E3 ligase Siah2 as well as the signaling platform molecule POSH (SH3RF1) conferred robust caspase-8 activation in response to TRAIL stimulus. Silencing Siah2 or POSH in prostate cancer cells led to increased caspase activity and apoptosis in response to both TRAIL and Fas ligand. The E3 activity of Siah2 was responsible for mediating apoptosis resistance; while POSH protein levels were critical for maintaining viability. Further characterization of Siah2 revealed it to function downstream of early death receptor events in the apoptotic pathway. The observed apoptosis resistance provides one biological explanation for the induction of Siah2 and POSH reported in lung and prostate cancer, respectively. Expanding on an initial yeast-two-hybrid screen we have confirmed a physical interaction between E3 ligases Siah2 and POSH. Utilizing a yeast-two-hybrid mapping approach we have defined the spacer region of POSH, more specifically the RPxAxVxP motif encompassing amino acids 601-607, to be the site of Siah2 binding. CONCLUSIONS: The data presented here define POSH and Siah2 as important mediators of death receptor mediated apoptosis and suggest targeting the interaction of these two E3 ligases is a promising novel cancer therapeutic strategy.


Subject(s)
Apoptosis , Nuclear Proteins/metabolism , Receptors, Death Domain/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Caspase 8/metabolism , Cell Survival , Gene Silencing , Humans , Molecular Sequence Data , Nuclear Proteins/genetics , Protein Binding , TNF-Related Apoptosis-Inducing Ligand/metabolism , Ubiquitin-Protein Ligases/genetics
5.
Eur Urol ; 59(5): 734-44, 2011 May.
Article in English | MEDLINE | ID: mdl-21269758

ABSTRACT

BACKGROUND: Quinazoline-based α1-adrenoceptor antagonists suppress tumor growth by inducing apoptosis via an α1-adrenoceptor-independent action. Anoikis is a unique mode of apoptosis consequential to insufficient cell-matrix interactions. OBJECTIVE: This study investigated the apoptotic effect of novel quinazoline-based compounds on human renal cancer cells. DESIGN, SETTING, AND PARTICIPANTS: Two cell lines were used: renal cell carcinoma (RCC) 786-0, harboring a von Hippel-Lindau (VHL) tumor-suppressor gene mutation with a highly angiogenic phenotype, and Caki cells (no VHL mutation). MEASUREMENTS: The lead compound DZ-50 (10 µM) led to significant inhibition of tumor-cell adhesion, migration, and invasion at a lower dose than doxazosin (25 µM) in both RCC lines. RESULTS AND LIMITATIONS: Doxazosin induced death-receptor-mediated apoptosis, while DZ-50 led to anoikis via targeting of the focal adhesion complex and AKT signaling that subsequently increased RCC susceptibility to caspase-8-mediated apoptosis. Both quinazoline compounds, doxazosin and DZ-50, significantly reduced RCC metastatic potential in vivo. CONCLUSIONS: Quinazoline-based drugs trigger anoikis in RCC by targeting the focal adhesion survival signaling. This potent antitumor action against human RCC suggests a novel quinazoline-based therapy targeting renal cancer.


Subject(s)
Anoikis/drug effects , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/enzymology , Doxazosin/analogs & derivatives , Focal Adhesion Kinase 1/metabolism , Kidney Neoplasms/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Caspase 8/metabolism , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Doxazosin/pharmacology , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mutation , Neoplasm Invasiveness , Receptors, Death Domain/drug effects , Receptors, Death Domain/metabolism , Time Factors , Transendothelial and Transepithelial Migration/drug effects , Von Hippel-Lindau Tumor Suppressor Protein/genetics
6.
Cell Stress Chaperones ; 15(5): 497-508, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20013084

ABSTRACT

IRE1 is a conserved dual endoribonuclease/protein kinase that is indispensable for directing the endoplasmic reticulum (ER) stress response in yeast, flies, and worms. In mammalian systems, however, the precise biological activities carried out by IRE1alpha are unclear. Here, molecular and chemical genetic approaches were used to control IRE1 activity in a number of prostate cancer cell lines and the resulting impact on gene transcription, cell survival, and proliferation was examined. Modulating IRE1alpha activity had no transcriptional effect on the induction of genes classically associated with the ER stress response (Grp78 and CHOP) or cell survival when confronted with ER stress agents. Rather, IRE1alpha activity was positively correlated to proliferation. Since Xbp-1 mRNA is the sole known substrate for IRE1 endoribonuclease activity, the role of this transcription factor in mediating proliferation was examined. Repressing total Xbp-1 levels by siRNA techniques effectively slowed proliferation. In an effort to identify IRE1/XBP-1 targets responsible for the cell cycle response, genome-wide differential mRNA expression analysis was performed. Consistent with its ability to sense ER stress, IRE1alpha induction led to an enrichment of ER-Golgi, plasma membrane, and secretory gene products. An increase in cyclin A1 expression was the only differentially expressed cell cycle regulatory gene found. Greater cyclin A protein levels were consistently observed in cells with active IRE1alpha and were dependent on XBP-1. We conclude that IRE1alpha activity controls a subset of the ER stress response and mediates proliferation through tight control of Xbp-1 splicing.


Subject(s)
Cyclin A1/metabolism , DNA-Binding Proteins/metabolism , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Cell Survival/physiology , Cyclin A1/genetics , DNA-Binding Proteins/genetics , Endoplasmic Reticulum Chaperone BiP , Endoribonucleases/genetics , Humans , Male , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/genetics , Regulatory Factor X Transcription Factors , Transcription Factors/genetics , X-Box Binding Protein 1
7.
Cell ; 138(2): 377-88, 2009 Jul 23.
Article in English | MEDLINE | ID: mdl-19632185

ABSTRACT

Prostate apoptosis response-4 (Par-4) is a proapoptotic protein with intracellular functions in the cytoplasm and nucleus. Unexpectedly, we noted Par-4 protein is spontaneously secreted by normal and cancer cells in culture, and by Par-4 transgenic mice that are resistant to spontaneous tumors. Short exposure to endoplasmic reticulum (ER) stress-inducing agents further increased cellular secretion of Par-4 by a brefeldin A-sensitive pathway. Secretion occurred independently of caspase activation and apoptosis. Interestingly, extracellular Par-4 induced apoptosis by binding to the stress response protein, glucose-regulated protein-78 (GRP78), expressed at the surface of cancer cells. The interaction of extracellular Par-4 and cell surface GRP78 led to apoptosis via ER stress and activation of the FADD/caspase-8/caspase-3 pathway. Moreover, apoptosis inducible by TRAIL, which also exerts cancer cell-specific effects, is dependent on extracellular Par-4 signaling via cell surface GRP78. Thus, Par-4 activates an extrinsic pathway involving cell surface GRP78 receptor for induction of apoptosis.


Subject(s)
Apoptosis , Receptors, Thrombin/metabolism , Animals , Brefeldin A/pharmacology , Cell Line , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Humans , Mice , Mice, Transgenic , Protein Structure, Tertiary , Protein Transport/drug effects , Receptors, Thrombin/chemistry
8.
Cancer Biol Ther ; 8(1): 73-80, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19029816

ABSTRACT

Inducing apoptosis via the extrinsic death receptor pathway is an attractive anti-cancer treatment strategy, however, numerous cancer cells exhibit significant resistance to death ligand stimuli. Here, we investigated the anti-neoplastic capability of proteasome inhibition, through the administration of Velcade, to synergize with a death receptor agonist in vivo. The death ligand-resistant LNCaP prostate xenograft model was utilized. Tumors were established and mice were treated with Velcade, TRAIL (TNF-Related Apoptosis Inducing Ligand) or the combined regimen. Only mice treated with a combination of Velcade and TRAIL was tumor growth inhibited with a corresponding loss of the hemorrhagic phenotype, decreased tumor cell proliferation and increased tumor cell apoptosis. Next, to determine if the extrinsic pathway is critical for mediating the anti-tumor efficacy that can be achieved in some cell types with Velcade treatment alone, the death receptor sensitive PC-3 xenograft model was used. PC-3 tumors exhibited a 54% decrease in tumor volume in response to Velcade, while c-FLIP overexpressing PC-3 xenografts were resistant to the treatment. These findings suggest that the extrinsic apoptotic pathway can mediate the anti-tumor effects of Velcade and support the therapeutic use of proteasome inhibition in combination with a death receptor stimulus in the treatment of prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Boronic Acids/pharmacology , Cell Division/drug effects , Prostatic Neoplasms/pathology , Pyrazines/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , Boronic Acids/therapeutic use , Bortezomib , Cell Line, Tumor , Humans , In Situ Nick-End Labeling , Male , Mice , Prostatic Neoplasms/drug therapy , Pyrazines/therapeutic use , Recombinant Proteins/pharmacology , TNF-Related Apoptosis-Inducing Ligand/therapeutic use
9.
J Urol ; 180(6): 2681-7, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18951569

ABSTRACT

PURPOSE: We evaluated gene expression profiles after inducing differentiation in cultured interstitial cystitis and control urothelial cells. MATERIALS AND METHODS: Bladder biopsies were taken from patients with interstitial cystitis and controls, that is women undergoing surgery for stress incontinence. Primary cultures were grown in keratinocyte growth medium with supplements. To induce differentiation in some plates the medium was changed to Dulbecco's modified Eagle's minimal essential medium-F12 (Media Tech, Herndon, Virginia) with supplements. RNA was analyzed with Affymetrix(R) chips. Three patients with nonulcerative interstitial cystitis were compared with 3 controls. RESULTS: After inducing differentiation 302 genes with a described function were altered at least 3-fold in interstitial cystitis and control cells (p <0.01). Functions of the 162 up-regulated genes included cell adhesion (eg claudins, occludin and cingulin), urothelial differentiation, the retinoic acid pathway and keratinocyte differentiation (eg skin cornified envelope components). The 140 down-regulated transcripts included genes associated with basal urothelium (eg p63, integrins beta4, alpha5 and alpha6, basonuclin 1 and extracellular matrix components), vimentin, metallothioneins, and members of the Wnt and Notch pathways. When comparing interstitial cystitis control cells after differentiation, only 7 genes with a described function were altered at least 3-fold (p <0.01). PI3, SERPINB4, CYP2C8, EFEMP2 and SEPP1 were decreased, and AKR1C2 and MKNK1 were increased in interstitial cystitis cases. CONCLUSIONS: Differentiation associated changes occurred in interstitial cystitis and control cells. Comparing interstitial cystitis vs control cases revealed few differences. This study may have included patients with interstitial cystitis and minimal urothelial deficiency, and/or we may have selected cells that were most robust in culture. Also, the abnormal urothelium in interstitial cystitis cases may be due to post-translational changes and/or to the bladder environment.


Subject(s)
Cell Differentiation/genetics , Cystitis, Interstitial/genetics , Cystitis, Interstitial/pathology , Gene Expression Regulation , Urinary Bladder/cytology , Urinary Bladder/pathology , Adult , Cells, Cultured , Female , Humans , Middle Aged , Urothelium/cytology
10.
Prostate ; 68(15): 1615-23, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18663729

ABSTRACT

Molecular targeting of apoptotic signaling pathways has been extensively studied in recent years and directed towards the development of effective therapeutic modalities for treating advanced androgen-independent prostate tumors. The majority of therapeutic agents act through intrinsic or mitochondrial pathways to induce programmed cell death. The induction of apoptosis through endoplasmic reticulum (ER) stress pathways may provide an alternative to treat patients. The functional interaction between the BCL-2 family members and regulation of calcium homeostasis in the ER provides a critical link to the life or death outcome of the cell. Apoptosis induction mediated by ER stress-inducing agents is just beginning to be exploited for therapeutic targeting of prostate tumors. Insightful dissection of recently discovered apoptotic signaling pathways that function through the endoplasmic reticulum may identify novel molecules that could effectively target both androgen-dependent and androgen-independent prostate tumors. In this review, we focus on linking ER stress-induced apoptosis to therapeutic targeting of prostate tumors and dissect its cross-talk with the intrinsic and extrinsic apoptotic pathways.


Subject(s)
Apoptosis , Endoplasmic Reticulum/metabolism , Prostatic Neoplasms/drug therapy , Signal Transduction , Stress, Physiological/physiopathology , Calcium/metabolism , Homeostasis , Humans , Male , Proto-Oncogene Proteins c-bcl-2/metabolism
11.
Prostate ; 68(2): 200-9, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18076022

ABSTRACT

BACKGROUND: Proteasome inhibition through the administration of Velcade is a viable chemotherapeutic strategy that is approved to treat multiple myeloma and is being evaluated for efficacy against prostate cancer. Currently, the apoptotic pathways that contribute to this anticancer response are poorly understood. Our goal is to test the extent to which proteasome inhibition modulates apoptosis through death receptor pathways. METHODS: Several prostate cancer cell lines and primary prostate epithelial cells (PrECs) were used as models. The death receptor pathway was activated by the expression of Fas ligand (FasL) or addition of TNF-related apoptosis-inducing ligand (TRAIL) in the presence or absence of proteasome inhibitors. The apoptotic response was quantified by annexin V, TUNEL and nuclear condensation assays. Western blot analysis was conducted to quantify protein levels and enzyme assays were used to measure caspase activity. RESULTS: Proteasome inhibition markedly sensitized prostate cancer cells to apoptosis initiated by Fas ligand (FasL) or TRAIL. In the presence of either death ligand, procaspase-8 processing occurred, but led to minimal amounts of active caspase-8. The addition of Velcade, however, led to robust active caspase-8 protein abundance and activity. In the presence of Velcade the caspase-8 p18 subunit half-life increased from 22 min to over 2 hr. CONCLUSIONS: These findings demonstrate that proteasome inhibition can sensitize cells to apoptosis elicited by tumor necrosis factor ligands and retarding caspase-8 degradation provides one explanation for this activity. This study suggests that the clinical efficacy of Velcade may result, at least in part, from the activity of death ligands.


Subject(s)
Apoptosis/physiology , Caspase 8/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteasome Inhibitors , Receptors, Death Domain/metabolism , Apoptosis/drug effects , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Cell Survival , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fas Ligand Protein/metabolism , Half-Life , Humans , Male , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology
12.
J Mol Biol ; 363(4): 813-22, 2006 Nov 03.
Article in English | MEDLINE | ID: mdl-16987528

ABSTRACT

The chemokine CXCL14/BRAK participates in immune surveillance by recruiting dendritic cells. CXCL14 gene expression is altered in a number of cancers, but protein expression levels have not been investigated. Here we report that CXCL14 protein can be expressed in primary epithelial cells; however, in several immortalized and cancer cell lines this protein is targeted for polyubiquitylation and proteasomal degradation. We determined the NMR structure of CXCL14 to identify motifs controlling its expression. CXCL14 adopts the canonical chemokine tertiary fold but contains a unique five amino acid insertion (41VSRYR45) relative to other CXC chemokines. Deletion or substitution of key residues within this insertion prevented proteasomal degradation. Furthermore, we defined a 15 amino acid fragment of CXCL14 that is sufficient to induce proteasomal degradation. This study elucidates a post-translational mechanism for the loss of CXCL14 in cancer and a novel mode of chemokine regulation.


Subject(s)
Chemokines, CXC/chemistry , Chemokines, CXC/metabolism , Proteasome Endopeptidase Complex/metabolism , Amino Acid Sequence , Enzyme Inhibitors/pharmacology , Humans , Interleukin-8/chemistry , Molecular Sequence Data , Mutant Proteins/metabolism , Nuclear Magnetic Resonance, Biomolecular , Proteasome Endopeptidase Complex/chemistry , Proteasome Inhibitors , Protein Structure, Secondary , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
13.
Neoplasia ; 7(9): 816-23, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16229804

ABSTRACT

Classic mechanisms of tumor response to chemotherapy include apoptosis and mitotic catastrophe. Recent studies have suggested that cellular senescence, a terminal proliferation arrest seen in vitro, may be invoked during the exposure of cancer cells to chemotherapeutic agents. To identify markers associated specifically with the cellular senescence phenotype, we utilized expression data from cDNA microarray experiments identifying transcripts whose expression levels increased as human prostate epithelial cells progressed to senescence. When screened against other growth-inhibitory conditions, including quiescence and apoptosis, many of these transcripts were also upregulated, indicating that similar pathways occur between apoptosis and senescence. A senescent-like phenotype was then induced in several prostate cancer cell lines using 5-aza-2'-deoxycytidine, doxorubicin, or Docetaxel. Treatment with these agents resulted in a significant increase in the induction of senescence-specific genes when compared to nonsenescent conditions. The performance of the panel was improved with fluorescence-activated cell sorting using PKH26 to isolate nonproliferating, viable, drug-treated populations, indicating that a heterogeneous response occurs with chemotherapy. We have defined an RNA-based gene panel that characterizes the senescent phenotype induced in cancer cells by drug treatment. These data also indicate that a panel of genes, rather than one marker, needs to be utilized to identify senescence.


Subject(s)
Antineoplastic Agents/pharmacology , Cellular Senescence/genetics , Prostatic Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Apoptosis/genetics , Cell Line, Tumor , Docetaxel , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Gene Expression Regulation, Neoplastic , Humans , Male , Microarray Analysis , Phenotype , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Taxoids/pharmacology , Taxoids/therapeutic use
14.
Prostate ; 64(1): 67-74, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15651028

ABSTRACT

BACKGROUND: Recent studies suggest inflammatory processes may be involved in the development or progression of prostate cancer. Chemokines are a family of cytokines that can play several roles in cancer progression including angiogenesis, inflammation, cell recruitment, and migration. METHODS: Real-time quantitative RT-PCR, in situ RNA hybridization, laser capture microscopy, immunohistochemistry, and cDNA array based technologies were used to examine CXCL14 (BRAK) expression in paired normal and tumor prostate. To determine the role CXCL14 expression has on cancer progression, LAPC4 cells were engineered to overexpress mouse or human CXCL14, and xenograft studies were performed. RESULTS: CXCL14 RNA expression was observed in normal and tumor prostate epithelium and focally in stromal cells adjacent to cancer. CXCL14 mRNA was significantly upregulated in localized prostate cancer and positively correlated with Gleason score. CXCL14 levels were unchanged in BPH specimens. LAPC4 cells expressing CXCL14 resulted in a 43% tumor growth inhibition (P = 0.019) in vivo compared to vector only xenografts. CONCLUSIONS: CXCL14 mRNA upregulation is a common feature in prostate cancer. The finding that CXCL14 expression inhibits tumor growth suggests this gene has tumor suppressive functions.


Subject(s)
Chemokines, CXC/genetics , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/physiopathology , Animals , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Prostate/physiology , RNA, Messenger/analysis
15.
J Biol Chem ; 279(50): 52218-26, 2004 Dec 10.
Article in English | MEDLINE | ID: mdl-15471867

ABSTRACT

The imprinted insulin-like growth factor-2 (IGF2) gene is an auto/paracrine growth factor expressed only from the paternal allele in adult tissues. In tissues susceptible to aging-related cancers, including the prostate, a relaxation of IGF2 imprinting is found, suggesting a permissive role for epigenetic alterations in cancer development. To determine whether IGF2 imprinting is altered in cellular aging and senescence, human prostate epithelial and urothelial cells were passaged serially in culture to senescence. Allelic analyses using an IGF2 polymorphism demonstrated a complete conversion of the IGF2 imprint status from monoallelic to biallelic, in which the development of senescence was associated with a 10-fold increase in IGF2 expression. As a mechanism, a 2-fold decrease in the binding of the enhancer-blocking element CCCTC-binding factor (CTCF) within the intergenic IGF2-H19 region was found to underlie this switch to biallelic IGF2 expression in senescent cells. This decrease in CTCF binding was associated with reduced CTCF expression in senescent cells. No de novo increases in methylation at the IGF2 CTCF binding site were seen. The forced down-regulation of CTCF expression using small interfering RNA in imprinted prostate cell lines resulted in an increase in IGF2 expression and a relaxation of imprinting. Our data suggest a novel mechanism for IGF2 imprinting regulation, that is, the reduction of CTCF expression in the control of IGF2 imprinting. We also demonstrate that altered imprinting patterns contribute to changes in gene expression in aging cells.


Subject(s)
Cellular Senescence/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genomic Imprinting , Insulin-Like Growth Factor II/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Base Sequence , CCCTC-Binding Factor , Cells, Cultured , DNA Methylation , DNA, Complementary/genetics , Down-Regulation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Male , Prostate/cytology , Prostate/metabolism , RNA, Long Noncoding , RNA, Small Interfering/genetics , RNA, Untranslated/genetics , Transfection , Urothelium/cytology , Urothelium/metabolism
16.
Cancer Res ; 63(15): 4614-9, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12907640

ABSTRACT

Cdc37 is a co-chaperone protein that recruits several immature client kinases to Hsp90 for proper folding. Cdc37 up-regulation is a common early event in localized human prostate cancer. Although targeted overexpression in mice leads to prostate epithelial cell hyperplasia, the effect of Cdc37 dysregulation in human prostate cells is unclear. In this study, we examine the role of Cdc37 in the growth regulation of normal prostate epithelial cells using a unique human model system. We demonstrate that Cdc37 overexpression drives proliferation, whereas loss of Cdc37 function arrests growth and leads to apoptosis. With increased Cdc37 expression, molecular analysis of Cdc37 client pathways demonstrates enhanced Raf-1 activity, greater Cdk4 levels, and reduced expression of the cyclin-dependent kinase inhibitor p16/CDKN2. To further investigate these downstream pathways, enhanced Raf-1 or Cdk4 activities were selectively induced in human prostate epithelial cells. Raf-1 activation inhibited proliferation and generated an enlarged, flattened morphology. Induction of Cdk4 activity using cyclin D1 overexpression, however, was sufficient to promote proliferation. These data indicate that Cdc37 induces proliferation and is critical for survival in human prostate epithelial cells. These alterations in cell division and survival may be important in the development and progression of early prostate cancer.


Subject(s)
Cell Cycle Proteins/physiology , Drosophila Proteins , Molecular Chaperones/physiology , Prostate/cytology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins , Apoptosis/physiology , Bromodeoxyuridine/pharmacokinetics , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Division/physiology , Cell Survival/physiology , Chaperonins , Cyclin D1/biosynthesis , Cyclin D1/genetics , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/metabolism , Disease Progression , Enzyme Activation , Epithelial Cells/metabolism , Epithelial Cells/physiology , Humans , Male , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/biosynthesis , Molecular Chaperones/genetics , Prostate/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism
17.
Cancer Lett ; 185(2): 191-9, 2002 Nov 28.
Article in English | MEDLINE | ID: mdl-12169393

ABSTRACT

We examined the status of a cell cycle checkpoint by immunohistochemically staining for p16 and pRb using multiple tissue arrays generated from 49 primary and 23 hormone-sensitive metastatic human prostate cancers. We find that p16, a cell cycle inhibitor, is paradoxically overexpressed in 83% of proliferating primary prostate cancers and increased expression correlates with a more rapid treatment failure (P=0.01) and a higher histologic grade (P=0.001). pRb staining is heterogeneous, loses expression infrequently (19%), and does not correlate with p16 expression. Loss of either p16 or pRb expression is found significantly (P=0.01) more commonly (55%) in metastatic specimens. The remarkable frequency of p16/pRb alterations and strong clinical associations implicates inactivation of this pathway as a critical determinant in prostate cancer progression.


Subject(s)
Adenocarcinoma/pathology , Cyclin-Dependent Kinase Inhibitor p16/analysis , Genes, Retinoblastoma , Genes, p16 , Neoplasm Proteins/analysis , Prostatic Neoplasms/pathology , Retinoblastoma Protein/analysis , Adenocarcinoma/chemistry , Adenocarcinoma/genetics , Aged , Disease Progression , Disease-Free Survival , G1 Phase , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Loss of Heterozygosity , Male , Middle Aged , Neoplasm Metastasis , Prostatic Neoplasms/chemistry , Prostatic Neoplasms/genetics
18.
J Biol Chem ; 277(17): 14877-83, 2002 Apr 26.
Article in English | MEDLINE | ID: mdl-11836256

ABSTRACT

Cellular senescence forms a barrier that inhibits the acquisition of an immortal phenotype, a critical feature in tumorigenesis. The inactivation of multiple pathways that positively regulate senescence are required for immortalization. To identify these pathways in an unbiased manner, we performed DNA microarray analyses to assess the expression of 20,000 genes in human prostate epithelial cells (HPECs) passaged to senescence. These gene expression patterns were then compared with those of HPECs immortalized with the human Papillomavirus 16 E7 oncoprotein. Senescent cells display gene expression patterns that reflect their nonproliferative, differentiated phenotype and express secretory proteases and extracellular matrix components. A comparison of genes transcriptionally up-regulated in senescence to those in which expression is significantly down-regulated in immortalized HPECs identified three genes: the chemokine BRAK, DOC1, and a member of the insulin-like growth factor axis, IGFBP-3. Expression of these genes is found to be uniformly lost in human prostate cancer cell lines and xenografts, and previously, their inactivation was documented in tumor samples. Thus, these genes may function in novel pathways that regulate senescence and are inactivated during immortalization. These changes may be critical not only in allowing cells to bypass senescence in vitro but in the progression of prostate cancer in vivo.


Subject(s)
Cellular Senescence , Gene Expression Profiling , Prostate/cytology , Aged , Cell Line, Transformed , Cell Transformation, Viral , Epithelial Cells/cytology , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Papillomaviridae/physiology , Prostate/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...