Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Curr Nutr Rep ; 11(4): 643-652, 2022 12.
Article in English | MEDLINE | ID: mdl-36018501

ABSTRACT

PURPOSE OF REVIEW: An epidemic of age-associated cognitive decline, most commonly ascribed to neurodegenerative conditions such as Alzheimer's and Parkinson's disease, is causing healthcare costs to soar and devastating caregivers. An estimated 6.5 million Americans are living today with Alzheimer's disease, with 13.8 million cases projected by mid-century. Although genetic mutations are known to cause neurodegeneration, autosomal dominant disease is very rare and most sporadic cases can be attributed, at least in part, to modifiable risk factors. RECENT FINDINGS: Diet is a potential modifiable risk factor in cognitive decline. Food communicates with the brain through a complex signaling web involving multiple cells, mediators and receptors. Gut-brain communication is modulated by microorganisms including bacteria, archaea, viruses, and unicellular eukaryotes, which together constitute the microbiota. Microbes not only play major roles in the digestion and fermentation of the food, providing nutrients and bioactive metabolites, but also reflect the type and amount of food consumed and food-borne toxic exposures. Food components modify the diversity and abundance of the microbial populations, maintain the integrity of the gut barrier, and regulate the passage of microbes and their metabolites into the blood stream where they modulate the immune system and communicate with body systems including the brain. This paper will focus on selected mechanisms through which interactions between diet and the gut microbiota can modify brain integrity and cognitive function with emphasis on the pathogenesis of the most common dementia, Alzheimer's disease.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Gastrointestinal Microbiome , Humans , Gastrointestinal Microbiome/physiology , Diet , Brain/metabolism
2.
Cancer Med ; 7(7): 3385-3392, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29905005

ABSTRACT

Development of neuroendocrine prostate cancer (NEPC) is emerging as a major problem in clinical management of advanced prostate cancer (PCa). As increasingly potent androgen receptor (AR)-targeting antiandrogens are more widely used, PCa transdifferentiation into AR-independent NEPC as a mechanism of treatment resistance becomes more common and precarious, since NEPC is a lethal PCa subtype urgently requiring effective therapy. Reprogrammed glucose metabolism of cancers, that is elevated aerobic glycolysis involving increased lactic acid production/secretion, plays a key role in multiple cancer-promoting processes and has been implicated in therapeutics development. Here, we examined NEPC glucose metabolism using our unique panel of patient-derived xenograft PCa models and patient tumors. By calculating metabolic pathway scores using gene expression data, we found that elevated glycolysis coupled to increased lactic acid production/secretion is an important metabolic feature of NEPC. Specific inhibition of expression of MCT4 (a plasma membrane lactic acid transporter) by antisense oligonucleotides led to reduced lactic acid secretion as well as reduced glucose metabolism and NEPC cell proliferation. Taken together, our results indicate that elevated glycolysis coupled to excessive MCT4-mediated lactic acid secretion is clinically relevant and functionally important to NEPC. Inhibition of MCT4 expression appears to be a promising therapeutic strategy for NEPC.

3.
Oncotarget ; 8(16): 25928-25941, 2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28460430

ABSTRACT

To avoid over- or under-treatment of primary prostate tumours, there is a critical need for molecular signatures to discriminate indolent from aggressive, lethal disease. Reprogrammed energy metabolism is an important hallmark of cancer, and abnormal metabolic characteristics of cancers have been implicated as potential diagnostic/prognostic signatures. While genomic and transcriptomic heterogeneity of prostate cancer is well documented and associated with tumour progression, less is known about metabolic heterogeneity of the disease. Using a panel of high fidelity patient-derived xenograft (PDX) models derived from hormone-naïve prostate cancer, we demonstrated heterogeneity of expression of genes involved in cellular energetics and macromolecular biosynthesis. Such heterogeneity was also observed in clinical, treatment-naïve prostate cancers by analyzing the transcriptome sequencing data. Importantly, a metabolic gene signature of increased one-carbon metabolism or decreased proline degradation was identified to be associated with significantly decreased biochemical disease-free patient survival. These results suggest that metabolic heterogeneity of hormone-naïve prostate cancer is of biological and clinical importance and motivate further studies to determine the heterogeneity in metabolic flux in the disease that may lead to identification of new signatures for tumour/patient stratification and the development of new strategies and targets for therapy of prostate cancer.


Subject(s)
Energy Metabolism/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Cluster Analysis , Disease Models, Animal , Gene Expression Profiling , Heterografts , High-Throughput Nucleotide Sequencing , Humans , Male , Metabolic Networks and Pathways , Mice , Prognosis , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Transcriptome
4.
EMBO Mol Med ; 8(7): 761-78, 2016 07.
Article in English | MEDLINE | ID: mdl-27198502

ABSTRACT

Clusterin (CLU) is a stress-activated molecular chaperone that confers treatment resistance to taxanes when highly expressed. While CLU inhibition potentiates activity of taxanes and other anti-cancer therapies in preclinical models, progression to treatment-resistant disease still occurs implicating additional compensatory survival mechanisms. Taxanes are believed to selectively target cells in mitosis, a complex mechanism controlled in part by balancing antagonistic roles of Cdc25C and Wee1 in mitosis progression. Our data indicate that CLU silencing induces a constitutive activation of Cdc25C, which delays mitotic exit and hence sensitizes cancer cells to mitotic-targeting agents such as taxanes. Unchecked Cdc25C activation leads to mitotic catastrophe and cell death unless cells up-regulate protective mechanisms mediated through the cell cycle regulators Wee1 and Cdk1. In this study, we show that CLU silencing induces a constitutive activation of Cdc25C via the phosphatase PP2A leading to relief of negative feedback inhibition and activation of Wee1-Cdk1 to promote survival and limit therapeutic efficacy. Simultaneous inhibition of CLU-regulated cell cycle effector Wee1 may improve synergistic responses of biologically rational combinatorial regimens using taxanes and CLU inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged-Ring Compounds/pharmacology , Cell Proliferation/drug effects , Clusterin/metabolism , Mitosis/drug effects , Prostatic Neoplasms/pathology , Taxoids/pharmacology , Cell Line, Tumor , Clusterin/genetics , Gene Knockdown Techniques , Humans , Male
5.
Neoplasia ; 17(11): 805-16, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26678908

ABSTRACT

Vascular endothelial growth factor (VEGF)-targeted antiangiogenic therapy significantly inhibits the growth of clear cell renal cell carcinoma (RCC). Eventually, therapy resistance develops in even the most responsive cases, but the mechanisms of resistance remain unclear. Herein, we developed two tumor models derived from an RCC cell line by conditioning the parental cells to two different stresses caused by VEGF-targeted therapy (sunitinib exposure and hypoxia) to investigate the mechanism of resistance to such therapy in RCC. Sunitinib-conditioned Caki-1 cells in vitro did not show resistance to sunitinib compared with parental cells, but when tested in vivo, these cells appeared to be highly resistant to sunitinib treatment. Hypoxia-conditioned Caki-1 cells are more resistant to hypoxia and have increased vascularity due to the upregulation of VEGF production; however, they did not develop sunitinib resistance either in vitro or in vivo. Human endothelial cells were more proliferative and showed increased tube formation in conditioned media from sunitinib-conditioned Caki-1 cells compared with parental cells. Gene expression profiling using RNA microarrays revealed that several genes related to tissue development and remodeling, including the development and migration of endothelial cells, were upregulated in sunitinib-conditioned Caki-1 cells compared with parental and hypoxia-conditioned cells. These findings suggest that evasive resistance to VEGF-targeted therapy is acquired by activation of VEGF-independent angiogenesis pathways induced through interactions with VEGF-targeted drugs, but not by hypoxia. These results emphasize that increased inhibition of tumor angiogenesis is required to delay the development of resistance to antiangiogenic therapy and maintain the therapeutic response in RCC.


Subject(s)
Angiogenesis Inhibitors/metabolism , Carcinoma, Renal Cell/metabolism , Drug Delivery Systems , Indoles/metabolism , Kidney Neoplasms/metabolism , Pyrroles/metabolism , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/administration & dosage , Animals , Carcinoma, Renal Cell/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Drug Delivery Systems/methods , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Indoles/administration & dosage , Kidney Neoplasms/drug therapy , Mice , Mice, Nude , Pyrroles/administration & dosage , Sunitinib , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Xenograft Model Antitumor Assays/methods
6.
Oncotarget ; 6(33): 34818-30, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26472187

ABSTRACT

Tumor microenvironments are characterized by decreased oxygen and nutrition due to the rapid and progressive nature of tumors and also stresses induced by several anti-tumor therapies. These intense cell stressors trigger a protective cell survival mechanism heralded by the unfolded protein response (UPR). The UPR is induced by an accumulation of unfolded proteins in the endoplasmic reticulum (ER) following cell starvation. Although the ER stress response is implicated in cytoprotection, its precise role during anti-angiogenic therapy remains unclear. One of the major proteins involved in ER stress is glucose-regulated protein 78 (GRP78), which binds to unfolded proteins and dissociates from membrane-bound ER stress sensors. To determine the role of ER stress responses during anti-angiogenic therapy and the potential role of GRP78 in combined therapy in renal cell carcinoma (RCC), we used GRP78 overexpressing or knockdown RCC cells under hypoxic or hypoglycemic conditions in vitro and in animal models treated with sunitinib. Here, we report that GRP78 plays a crucial role in protecting RCC cells from hypoxic and hypoglycemic stress induced by anti-angiogenic therapy. Knockdown of GRP78 using siRNA inhibited cancer cell survival and induced apoptosis in RCC cells in vitro and also resulted in ER stress-induced apoptosis and hypoxic/hypoglycemic stress-induced apoptosis by inactivating the PERK/eIF-2α pathway. Finally, GRP78 knockdown showed potent suppression of tumor growth and enhanced the antitumor effect of sunitinib in RCC xenografts. Our findings suggest that GRP78 may serve as a novel therapeutic target in combination with anti-angiogenic therapy for the management of RCC.


Subject(s)
Carcinoma, Renal Cell/pathology , Endoplasmic Reticulum Stress/physiology , Heat-Shock Proteins/metabolism , Kidney Neoplasms/pathology , eIF-2 Kinase/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Blotting, Western , Cell Hypoxia/physiology , Cell Line, Tumor , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Female , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Immunohistochemistry , Indoles/pharmacology , Mice , Mice, Nude , Microscopy, Confocal , Pyrroles/pharmacology , RNA, Small Interfering , Signal Transduction/drug effects , Signal Transduction/physiology , Stress, Physiological , Sunitinib , Transfection , Xenograft Model Antitumor Assays
7.
Mol Cancer Ther ; 14(4): 1024-34, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25657336

ABSTRACT

Renal cell carcinoma (RCC) is the most common malignancy in the kidney. Antiangiogenic targeted therapies inhibit the progression of RCC, but have limited impacts on invasion or metastasis of tumor cells. Integrin-linked kinase (ILK) is a serine/threonine kinase implicated in the regulation of cell growth/survival, cell-cycle progression, epithelial-mesenchymal transition (EMT), invasion/migration, and angiogenesis. However, the role of ILK in RCC has not been evaluated. We investigated the role of ILK on cancer progression and metastasis and the therapeutic potential of ILK inhibition in RCC. Our investigation reveals that ILK is expressed at a low level in normal cells and low-stage RCC cells and is highly expressed in advanced and metastatic cells. Caki-1, a metastatic RCC cell line, showed higher expression of molecular EMT markers, including Snail and Zeb1, but decreased activity of GSK3ß. Knockdown of ILK using small interference (si)-ILK minimally inhibited tumor proliferation and cell-cycle progression was not significantly affected. However, ILK knockdown suppressed the formation of stress fibers and focal adhesions and impeded phenotypic EMT markers, including cell migration and invasion, in Caki-1 and UMRC-3 cells. Finally, in vivo knockdown of ILK suppressed the progression, invasion, and metastasis of primary RCC in nude mice by downregulation of EMT markers (Snail, Zeb1, vimentin, and E-cadherin). Our results show that ILK may be essential for invasion and metastasis in RCC and regulates vimentin and E-cadherin expression by regulating the EMT-related transcription factors Snail and Zeb1. These results suggest that ILK may be a potential target in RCC.


Subject(s)
Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Epithelial-Mesenchymal Transition/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Biomarkers , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Movement , Cell Survival/genetics , Disease Models, Animal , Down-Regulation , Gene Expression , Gene Knockdown Techniques , Heterografts , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Neoplasm Staging , Phenotype , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/genetics , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Finger E-box-Binding Homeobox 1
8.
J Am Anim Hosp Assoc ; 50(2): 96-104, 2014.
Article in English | MEDLINE | ID: mdl-24446402

ABSTRACT

An image-guided robotic stereotactic radiosurgery (SRS) system can be used to deliver curative-intent radiation in either single fraction or hypofractionated doses. Medical records for 19 dogs with nonlymphomatous nasal tumors treated with hypofractionated image-guided robotic stereotactic body radiotherapy (SBRT), either with or without adjunctive treatment, were retrospectively analyzed for survival and prognostic factors. Median survival time (MST) was evaluated using Kaplan-Meier survival curves. Age, breed, tumor type, stage, tumor size, prescribed radiation dose, and heterogeneity index were analyzed for prognostic significance. Dogs were treated with three consecutive-day, 8-12 gray (Gy) fractions of image-guided robotic SBRT. Overall MST was 399 days. No significant prognostic factors were identified. Acute side effects were rare and mild. Late side effects included one dog with an oronasal fistula and six dogs with seizures. In three of six dogs, seizures were a presenting complaint prior to SBRT. The cause of seizures in the remaining three dogs could not be definitively determined due to lack of follow-up computed tomography (CT) imaging. The seizures could have been related to either progression of disease or late radiation effect. Results indicate that image-guided robotic SBRT, either with or without adjunctive therapy, for canine nonlymphomatous nasal tumors provides comparable survival times (STs) to daily fractionated megavoltage radiation with fewer required fractions and fewer acute side effects.


Subject(s)
Dog Diseases/radiotherapy , Nose Neoplasms/veterinary , Radiotherapy, Image-Guided/veterinary , Animals , Dog Diseases/mortality , Dog Diseases/pathology , Dogs , Dose Fractionation, Radiation , Lymphatic Metastasis , Nose Neoplasms/radiotherapy , Retrospective Studies , Survival Analysis , Treatment Outcome
9.
J Urol ; 190(4): 1404-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23500642

ABSTRACT

PURPOSE: KU7 is a popular urothelial carcinoma cell line that was isolated from the bladder of a patient at Keio University in 1980. It has subsequently been widely used in laboratories around the world. We describe how routine cell line authentication revealed that KU7 was cross contaminated almost 30 years ago with HeLa, a cervical carcinoma cell line. MATERIALS AND METHODS: Presumed KU7 clones dating from 1984 to 1999 were provided by M.D. Anderson Cancer Center, Vancouver Prostate Centre, Kyoto University, Tokyo Medical University and Keio University. HeLa was obtained from ATCC. Genomic DNA was isolated and short tandem repeat analysis was performed at the M.D. Anderson Cancer Center Characterized Cell Line Core Facility, Johns Hopkins University Fragment Analysis Facility and RIKEN BioResource Center, Ibaraki, Japan. Comparative genomic hybridization was performed on a platform (Agilent Technologies, Santa Clara, California) at Vancouver Prostate Centre. RESULTS: The short tandem repeat profile of all KU7 clones was an exact match with that of HeLa. Comparative genomic hybridization of all samples revealed an abundance of shared chromosomal aberrations. Slight differences in some genomic areas were explained by genomic drift in different KU7 clones separated by many years. CONCLUSIONS: Our analysis identified that cross contamination of KU7 with HeLa occurred before 1984 at the source institution. All KU7 clones in the urological literature should be considered HeLa and experimental results should be viewed in this light. Our results emphasize the need to authenticate cell lines in oncological research.


Subject(s)
DNA Contamination , HeLa Cells , Comparative Genomic Hybridization , Gene Expression Profiling , Humans , Time Factors , Urinary Bladder Neoplasms/pathology
10.
Cancer Cell ; 23(3): 332-46, 2013 Mar 18.
Article in English | MEDLINE | ID: mdl-23518348

ABSTRACT

Understanding the mechanism underlying the regulation of the androgen receptor (AR), a central player in the development of castration-resistant prostate cancer (CRPC), holds promise for overcoming the challenge of treating CRPC. We demonstrate that the ubiquitin ligase Siah2 targets a select pool of NCOR1-bound, transcriptionally-inactive AR for ubiquitin-dependent degradation, thereby promoting expression of select AR target genes implicated in lipid metabolism, cell motility, and proliferation. Siah2 is required for prostate cancer cell growth under androgen-deprivation conditions in vitro and in vivo, and Siah2 inhibition promotes prostate cancer regression upon castration. Notably, Siah2 expression is markedly increased in human CRPCs. Collectively, we find that selective regulation of AR transcriptional activity by the ubiquitin ligase Siah2 is important for CRPC development.


Subject(s)
Nuclear Proteins/metabolism , Nuclear Receptor Co-Repressor 1/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Transcription, Genetic , Ubiquitin-Protein Ligases/metabolism , Animals , Castration , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Lipid Metabolism , Male , Mice , Nuclear Proteins/genetics , RNA Interference , RNA, Small Interfering , Signal Transduction , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics
11.
Endocr Relat Cancer ; 20(2): 173-86, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23319492

ABSTRACT

IGF2 is a mitogenic foetal growth factor commonly over-expressed in cancers, including prostate cancer (PC). We recently demonstrated that insulin can activate de novo steroidogenesis in PC cells, a major pathway for reactivation of androgen pathways and PC progression. IGF2 can activate the IGF1 receptor (IGF1R) or insulin receptor (INSR) or hybrids of these two receptors. We therefore hypothesized that IGF2 may contribute to PC progression via de novo steroidogenesis. IGF2 mRNA but not IGF2 receptor mRNA expression was increased in patient samples during progression to castrate-resistant PC as was immunoreactivity to INSR and IGF1R antibodies. Treatment of androgen receptor (AR)-positive PC cell lines LNCaP and 22RV1 with IGF2 for 48 h resulted in increased expression of steroidogenic enzyme mRNA and protein, including steroid acute regulatory protein (StAR), cytochrome p450 family member (CYP)17A1, aldo-keto reductase family member (AKR)1C3 and hydroxysteroid dehydrogenase (HSD)17B3. IGF2 treatment resulted in increased steady state steroid levels and increased de novo steroidogenesis resulting in AR activation as demonstrated by PSA mRNA induction. Inhibition of the IGF1R/INSR signalling axis attenuated the effects of IGF2 on steroid hormone synthesis. We present a potential mechanism for prostatic IGF2 contributing to PC progression by inducing steroidogenesis and that IGF2 signalling and related pathways present attractive targets for PC therapy.


Subject(s)
Insulin-Like Growth Factor II/pharmacology , Prostatic Neoplasms/metabolism , Steroids/biosynthesis , Cell Line, Tumor , Humans , Insulin-Like Growth Factor II/genetics , Male , Prostate-Specific Antigen/genetics , RNA, Messenger/metabolism , Receptor, IGF Type 2/genetics
12.
Mol Cancer Res ; 8(1): 119-30, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20068069

ABSTRACT

Secretory clusterin (sCLU) is a stress-activated, cytoprotective chaperone that confers broad-spectrum cancer treatment resistance, and its targeted inhibitor (OGX-011) is currently in phase II trials for prostate, lung, and breast cancer. However, the molecular mechanisms by which sCLU inhibits treatment-induced apoptosis in prostate cancer remain incompletely defined. We report that sCLU increases NF-kappaB nuclear translocation and transcriptional activity by serving as a ubiquitin-binding protein that enhances COMMD1 and I-kappaB proteasomal degradation by interacting with members of the SCF-betaTrCP E3 ligase family. Knockdown of sCLU in prostate cancer cells stabilizes COMMD1 and I-kappaB, thereby sequestrating NF-kappaB in the cytoplasm and decreasing NF-kappaB transcriptional activity. Comparative microarray profiling of sCLU-overexpressing and sCLU-knockdown prostate cancer cells confirmed that the expression of many NF-kappaB-regulated genes positively correlates with sCLU levels. We propose that elevated levels of sCLU promote prostate cancer cell survival by facilitating degradation of COMMD1 and I-kappaB, thereby activating the canonical NF-kappaB pathway.


Subject(s)
Carcinoma/metabolism , Carrier Proteins/metabolism , Clusterin/physiology , I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Prostatic Neoplasms/metabolism , Adaptor Proteins, Signal Transducing , Carcinoma/genetics , Carcinoma/pathology , Cell Line, Tumor , Clusterin/antagonists & inhibitors , Clusterin/genetics , Clusterin/metabolism , HeLa Cells , Humans , Male , Models, Biological , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , Protein Stability/drug effects , RNA, Small Interfering/pharmacology , Ubiquitin/metabolism , Ubiquitination/genetics
13.
Prostate ; 70(3): 239-51, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19790237

ABSTRACT

BACKGROUND: De novo androgen synthesis and subsequent androgen receptor (AR) activation has recently been shown to contribute to castration-resistant prostate cancer (CRPC) progression. Herein we provide evidence that fatty acids (FA) can trigger androgen synthesis within steroid starved prostate cancer (CaP) tumor cells. METHODS: Tumoral FA and steroid levels were assessed by GC-MS and LC-MS, respectively. Profiles of genes and proteins involved in FA activation of steroidogenesis were assessed by fluorescence microscopy, immunohistochemistry, microarray expression profiling and Western blot analysis. RESULTS: In human CaP tissues the levels of proteins responsible for FA activation of steroid synthesis were observed to be altered during progression to CRPC. Further investigating this mechanism in LNCaP cells, we demonstrate that specific FA, arachidonic acid, is synthesized in an androgen-dependent and AR-mediated manner. Arachidonic acid is known to induce steroidogenic acute regulatory protein (StAR) in steroidogenic cells. When bound to hormone sensitive lipase (HSL), StAR shuttles free cholesterol into the mitochondria for downstream conversion into androgens. We show that arachidonic acid induces androgen production in steroid starved LNCaP cells coincidently in the same conditions that HSL and StAR are predominantly localized in the mitochondria. Furthermore, their activities are verified by a functional increase in mitochondrial uptake of cholesterol in this steroid starved environment. CONCLUSIONS: We propose that this characterized arachidonic acid induced steroidogenesis mechanism significantly contributes to the activation of AR in CRPC progression and therefore recommend that fatty acid pathways be targeted therapeutically in progressing CaP.


Subject(s)
Androgen Antagonists/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Arachidonic Acid/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Steroids/biosynthesis , Androgens/deficiency , Androgens/metabolism , Animals , Arachidonic Acid/pharmacology , Cell Line, Tumor , Cholesterol/pharmacokinetics , Disease Progression , Drug Resistance , Humans , Male , Mice , Mice, Nude , Mitochondria/metabolism , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Phosphoproteins/metabolism , Prostatic Neoplasms/drug therapy , Sterol Esterase/metabolism , Tissue Distribution , Transplantation, Heterologous , Up-Regulation
14.
Clin Cancer Res ; 14(18): 5769-77, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18794086

ABSTRACT

PURPOSE: GLI transcription factors mediate hedgehog signaling and have been implicated in several human malignancies, including prostate cancer. The objectives of this study were to characterize GLI2 expression levels in human prostate cancer cell lines and tissues to test the effect of antisense oligonucleotide (ASO) targeting GLI2 on androgen-independent (AI) prostate cancer cell lines. EXPERIMENTAL DESIGN: A tissue microarray was used to characterize differences in GLI2 expression in benign prostate hyperplasia, prostate cancer treated by neoadjuvant hormonal therapy and AI prostate cancer. The effects of GLI2 ASO on PC-3 cell growth and paclitaxel chemosensitivity were assessed in vitro and in vivo. Oligonucleotide spotted microarray analysis was used to determine alteration in GLI2 coregulated genes after ASO treatment. RESULTS: The expression of GLI2 was significantly higher in prostate cancer than in benign prostate hyperplasia, decreased after androgen ablation in a time-dependent fashion, but became highly expressed again in AI prostate cancer. GLI2 ASO treatment of PC-3 cells reduced GLI2 mRNA and protein levels in a dose-dependent manner. GLI2 knockdown increased PC-3 cell apoptotic rates and significantly decreased cell growth and modulated levels of apoptosis-related genes, such as Bcl2, Bcl-xL, and clusterin. GLI2 knockdown also changed levels of several cell cycle regulators, such as cyclin D1, p27, and PKC-eta. Systematic administration of GLI2 ASO in athymic mice significantly delayed PC-3 tumor progression and enhanced paclitaxel chemosensitivity. CONCLUSIONS: These findings suggest that increased levels of GLI2 correlates with AI progression and that GLI2 may be a therapeutic target in castrate-resistant prostate cancer.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Nuclear Proteins/metabolism , Oligonucleotides, Antisense/pharmacology , Paclitaxel/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Androgens/pharmacology , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis , Base Sequence , Cell Line, Tumor , Disease Progression , Dose-Response Relationship, Drug , Humans , Kruppel-Like Transcription Factors/antagonists & inhibitors , Male , Membrane Proteins/analysis , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Nuclear Proteins/antagonists & inhibitors , Prostatic Neoplasms/genetics , Saccharomyces cerevisiae Proteins/analysis , Zinc Finger Protein Gli2
15.
Cancer Res ; 68(15): 6407-15, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18676866

ABSTRACT

Although systemic androgen deprivation prolongs life in advanced prostate cancer, remissions are temporary because patients almost uniformly progress to a state of a castration-resistant prostate cancer (CRPC) as indicated by recurring PSA. This complex process of progression does not seem to be stochastic as the timing and phenotype are highly predictable, including the observation that most androgen-regulated genes are reactivated despite castrate levels of serum androgens. Recent evidence indicates that intraprostatic levels of androgens remain moderately high following systemic androgen deprivation therapy, whereas the androgen receptor (AR) remains functional, and silencing the AR expression following castration suppresses tumor growth and blocks the expression of genes known to be regulated by androgens. From these observations, we hypothesized that CRPC progression is not independent of androgen-driven activity and that androgens may be synthesized de novo in CRPC tumors leading to AR activation. Using the LNCaP xenograft model, we showed that tumor androgens increase during CRPC progression in correlation to PSA up-regulation. We show here that all enzymes necessary for androgen synthesis are expressed in prostate cancer tumors and some seem to be up-regulated during CRPC progression. Using an ex vivo radiotracing assays coupled to high-performance liquid chromatography-radiometric/mass spectrometry detection, we show that tumor explants isolated from CRPC progression are capable of de novo conversion of [(14)C]acetic acid to dihydrotestosterone and uptake of [(3)H]progesterone allows detection of the production of six other steroids upstream of dihydrotestosterone. This evidence suggests that de novo androgen synthesis may be a driving mechanism leading to CRPC progression following castration.


Subject(s)
Androgens/metabolism , Orchiectomy , Prostatic Neoplasms/metabolism , Androgens/biosynthesis , Animals , Base Sequence , Blotting, Western , Cell Line, Tumor , Chromatography, High Pressure Liquid , Culture Media , DNA Primers , Disease Progression , Humans , Male , Mice , Mice, Nude , Polymerase Chain Reaction , Progesterone/administration & dosage , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Spectrometry, Mass, Electrospray Ionization
16.
Int J Cancer ; 122(10): 2368-76, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18240145

ABSTRACT

Therapeutic resistance is the underlying cause for most cancer deaths and a major problem associated with treatment of metastatic prostate cancer. HTI-286, a fully synthetic analog of the natural tripeptide hemiasterlin, inhibits tubulin polymerization and circumvents transport-based resistance to taxanes. In our study, we evaluated its inhibitory effects on human prostate cancer growth in vitro and in different in vivo models. Androgen-dependent and androgen-independent prostate cancer cell lines including a docetaxel-refractory PC-3 subline (PC-3dR) were treated with HTI-286. Transcriptional profiling was carried out to screen for changes in gene expression induced by HTI-286 and compared to docetaxel. In vivo, nude mice with established PC-3 or PC-3dR xenografts were given HTI-286 intravenously. Additionally, mice bearing hormone-sensitive LNCaP tumors were treated with castration in combination with early or delayed HTI-286 therapy. In all cell lines tested, HTI-286 was a potent inhibitor of proliferation and induced marked increases in apoptosis. Despite similar transcriptomic changes regarding cell death and cell cycle regulating genes after exposure to HTI-286 or docetaxel, array analysis revealed distinct molecular signatures for both compounds. Invivo, HTI-286 significantly inhibited growth of PC-3 and LNCaP xenografts and retained potency in PC-3dR tumors. Simultaneous castration plus HTI-286 therapy was superior to sequential treatment in the LNCaP model. In conclusion, HTI-286 showed strong antitumor activity both in androgen-dependent and androgen- independent tumors and may be a promising agent in second- line treatment strategies for patients suffering from docetaxel- refractory prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms, Hormone-Dependent/drug therapy , Oligopeptides/chemistry , Oligopeptides/pharmacology , Prostatic Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Docetaxel , Flow Cytometry , Gene Expression Profiling , Humans , Male , Mice , Mice, Nude , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Radiation-Sensitizing Agents/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Taxoids/pharmacology , Tumor Cells, Cultured/drug effects , Xenograft Model Antitumor Assays
17.
BJU Int ; 98(5): 1082-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16879439

ABSTRACT

OBJECTIVES: To evaluate synthetic small interference RNA (siRNA) compounds targeting heat-shock protein 27 (Hsp27) as an alternative approach to Hsp27 'knockdown' in prostate cancer cells, as Hsp27 expression is highly up-regulated in prostate cancer cells after androgen withdrawal or chemotherapy, to become uniformly highly expressed in androgen-independent (AI) prostate cancer. MATERIALS AND METHODS: We recently showed that targeting Hsp27 by a 2'-methoxyethyl modified phosphorothioate antisense oligonucleotide, OGX-427, inhibits Hsp27 expression and enhances hormone- and chemotherapy in prostate cancer xenograft models. In the present study, a 'gene walk' screening different siRNAs was initially used in PC-3 and LNCaP cells to determine the most potent sequence to down-regulate Hsp27 mRNA and protein levels. The effects of Hsp27 silencing on in vitro growth rates were studied by tetrazolium-blue and crystal violet assays. Apoptosis was determined by single-stranded DNA nuclear and cleaved caspase-3 immunostaining, as well as flow cytometry. Spotted microarrays with 14,000 human oligonucleotides were used to examine changes in gene expression. RESULTS: Low concentrations of 1 nm siRNA decreased Hsp27 mRNA levels by 19-fold and suppressed protein expression to undetectable levels. Silencing of Hsp27 in prostate cancer cells by siRNA # 2 increased apoptotic rates 2.4-4 fold and caused 40-76% inhibition of cell growth in LNCaP and PC-3 cells. Characteristic cleavage of caspase-3 occurred after treatment with Hsp27 siRNA (1 nm). cDNA microarray analysis from LNCaP and PC-3 cell lines revealed differential gene expression profiles after Hsp27 down-regulation that could be used to identify various survival pathways involved in androgen-dependent and AI growth. CONCLUSIONS: These findings illustrate the potential utility of Hsp27-silencing therapy and highlight Hsp27 siRNA strategies as a novel and highly effective tool, with the potential for future targeted therapy in enhancing the efficacy of chemotherapy in advanced prostate cancer.


Subject(s)
Caspase 3/metabolism , Heat-Shock Proteins/drug effects , Prostatic Neoplasms/drug therapy , RNA, Small Interfering/pharmacology , Apoptosis/drug effects , Blotting, Northern , Blotting, Western , Cell Line, Tumor , Down-Regulation/drug effects , Enzyme Activation , Flow Cytometry , Heat-Shock Proteins/metabolism , Humans , Male , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering/therapeutic use , Tumor Cells, Cultured , Up-Regulation/drug effects
18.
J Am Vet Med Assoc ; 228(7): 1053-62, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16579784

ABSTRACT

OBJECTIVE: To determine whether argyrophilic nucleolar organizing regions (AgNORs), Ki-67, and proliferating cell nuclear antigen (PCNA) scores were associated with histologic grade and survival in dogs with soft tissue sarcomas (STSs). DESIGN: Retrospective study. ANIMALS: 60 dogs with STSs. PROCEDURE: Medical records were examined and histologic specimens were reviewed. Tissue specimens obtained from archival materials were used to prepare sections for histologic staining for AgNOR and immunohistochemical staining for Ki-67 and PCNA labeling. Follow-up monitoring was obtained by reevaluation or telephone conversations with referring veterinarians or owners. RESULTS: 27 (45%) STSs were grade 1, 23 (38%) were grade 2, and 10 (17%) were grade 3. The mean and median AgNOR, Ki-67, and PCNA scores were determined, and significant positive associations among AgNOR and Ki-67 scores with histologic grade and mitotic score were detected. Fifty-four dogs had adequate follow-up examinations and were included in survival analysis and evaluation of prognostic factors. Overall median survival time was > 1,306 days. Twelve of 54 (22%) dogs died of tumor-related causes. Metastatic disease developed in 8 of 54 (15%) dogs. Results of univariate analysis indicated that increased mitotic score, increased AgNOR score, increased Ki-67 score, incomplete surgical margins, noncurative intent surgery, Ki-67 score greater than the median Ki-67 score, and AgNOR score greater than the median AgNOR score were prognostic factors for decreased survival time. Results of multivariate analysis indicated that increased AgNOR score was the only prognostic factor for decreased survival time. CONCLUSIONS AND CLINICAL RELEVANCE: Results suggested that AgNORs and possibly Ki-67 should be routinely evaluated with histologic grading for STSs in dogs.


Subject(s)
Antigens, Nuclear/metabolism , Dog Diseases/pathology , Ki-67 Antigen/metabolism , Neoplasm Staging/veterinary , Nuclear Proteins/metabolism , Sarcoma/veterinary , Soft Tissue Neoplasms/veterinary , Animals , Antigens, Nuclear/analysis , Dog Diseases/metabolism , Dog Diseases/mortality , Dogs , Female , Ki-67 Antigen/analysis , Male , Multivariate Analysis , Nuclear Proteins/analysis , Prognosis , Proliferating Cell Nuclear Antigen/analysis , Proliferating Cell Nuclear Antigen/metabolism , Retrospective Studies , Sarcoma/metabolism , Sarcoma/mortality , Sarcoma/pathology , Soft Tissue Neoplasms/metabolism , Soft Tissue Neoplasms/mortality , Soft Tissue Neoplasms/pathology , Survival Analysis
19.
Prostate ; 66(9): 971-86, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16541420

ABSTRACT

BACKGROUND: Despite evidence implicating insulin-like growth factor binding protein-3 (IGFBP-3) as a growth inhibitor of prostate cancer (CaP), little is known about changes in its regulation and function during progression to androgen independence. METHODS: The expression levels of IGFBP-3 were determined by cDNA microarray analysis and tissue microarrays (TMAs) after androgen ablations. LNCaP (LN-BP3) and C4-2 (C4-2-BP3) sublines were used to compare the apoptotic effects of IGFBP-3 in LNCaP (androgen-dependent) and C4-2 (androgen-independent) cells. RESULTS: After androgen deprivation, IGFBP-3 mRNA levels increased more in C4-2 compared to LNCaP cells. Androgens suppressed IGFBP-3 levels in a dose-dependent manner in LNCaP and C4-2 cell. IGFBP-3 expression was increased after NHT in human CaP tissues. Apoptotic rates increased in LN-BP3, but not C4-2-BP3 cells, following doxycycline-mediated IGFBP-3 induction. CONCLUSIONS: C4-2 cell survival in an androgen-depleted environment may be facilitated through differential resistance to the apoptotic effects elicited by IGFBP-3.


Subject(s)
Androgens/physiology , Gene Expression Regulation, Neoplastic/physiology , Insulin-Like Growth Factor Binding Protein 3/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/physiology , Androgens/pharmacology , Apoptosis/genetics , Apoptosis/physiology , Cell Line, Tumor , Cell Lineage , Cell Survival , Disease Progression , Dose-Response Relationship, Drug , Doxycycline/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Insulin-Like Growth Factor Binding Protein 3/physiology , Male , Neoplasms, Hormone-Dependent/chemistry , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/chemistry , RNA, Messenger/analysis , RNA, Messenger/genetics
20.
Cancer Res ; 65(23): 11083-93, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16322258

ABSTRACT

One strategy to improve therapies in prostate cancer involves targeting cytoprotective genes activated by androgen withdrawal to delay the emergence of the androgen-independent (AI) phenotype. The objectives of this study were to define changes in Hsp27 levels after androgen ablation and to evaluate the functional relevance of these changes in AI progression. Using a tissue microarray of 232 specimens of hormone-naïve and post-hormone ablation-treated prostate cancer, we found that Hsp27 levels increase after androgen ablation to become highly expressed (>4-fold, P < or = 0.01) in AI tumors. Hsp27 overexpression rendered LNCaP cells highly resistant to androgen withdrawal both in vitro and in vivo. Tumor volume and serum prostate-specific antigen levels increased 4.3- and 10-fold faster after castration when Hsp27 was overexpressed. Treatment of LNCaP tumor cells in vitro with Hsp27 antisense oligonucleotides (ASO) or short-interfering RNA suppressed Hsp27 levels in a dose-dependent and sequence-specific manner increased the apoptotic sub-G0-G1 fraction and caspase-3 cleavage >2-fold, as well as decreased signal transducers and activators of transcription 3 (Stat3) levels and its downstream genes, c-fos and sPLA-2. The cytoprotection afforded by Hsp27 overexpression was attenuated by Stat3 knockdown using specific Stat3 ASO. Coimmunoprecipitation and immunofluorescence confirmed that Hsp27 interacts with Stat3 and that Stat3 levels correlated directly with Hsp27 levels. Hsp27 ASO treatment in athymic mice bearing LNCaP tumors significantly delayed LNCaP tumor growth after castration, decreasing mean tumor volume and serum prostate-specific antigen levels by 57% and 69%, respectively. These findings identify Hsp27 as a modulator of Stat3-regulated apoptosis after androgen ablation and as a potential therapeutic target in advanced prostate cancer.


Subject(s)
Heat-Shock Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Prostatic Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Animals , Apoptosis/genetics , Caspase 3 , Caspases/metabolism , Cell Growth Processes/genetics , Cell Line, Tumor , Disease Progression , HSP27 Heat-Shock Proteins , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Male , Mice , Mice, Nude , Molecular Chaperones , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Neoplasms, Hormone-Dependent/surgery , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Orchiectomy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...