Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 30(10): 2272-2285, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38488813

ABSTRACT

PURPOSE: Develop and deploy a robust discovery platform that encompasses heterogeneity, clinical annotation, and molecular characterization and overcomes the limited availability of prostate cancer models. This initiative builds on the rich MD Anderson (MDA) prostate cancer (PCa) patient-derived xenograft (PDX) resource to complement existing publicly available databases by addressing gaps in clinically annotated models reflecting the heterogeneity of potentially lethal and lethal prostate cancer. EXPERIMENTAL DESIGN: We performed whole-genome, targeted, and RNA sequencing in representative samples of the same tumor from 44 PDXs derived from 38 patients linked to donor tumor metadata and corresponding organoids. The cohort includes models derived from different morphologic groups, disease states, and involved organ sites (including circulating tumor cells), as well as paired samples representing heterogeneity or stages before and after therapy. RESULTS: The cohort recapitulates clinically reported alterations in prostate cancer genes, providing a data resource for clinical and molecular interrogation of suitable experimental models. Paired samples displayed conserved molecular alteration profiles, suggesting the relevance of other regulatory mechanisms (e.g., epigenomic) influenced by the microenvironment and/or treatment. Transcriptomically, models were grouped on the basis of morphologic classification. DNA damage response-associated mechanisms emerged as differentially regulated between adenocarcinoma and neuroendocrine prostate cancer in a cross-interrogation of PDX/patient datasets. CONCLUSIONS: We addressed the gap in clinically relevant prostate cancer models through comprehensive molecular characterization of MDA PCa PDXs, providing a discovery platform that integrates with patient data and benchmarked to therapeutically relevant consensus clinical groupings. This unique resource supports robust hypothesis generation and testing from basic, translational, and clinical perspectives.


Subject(s)
Prostatic Neoplasms , Humans , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Male , Animals , Mice , Xenograft Model Antitumor Assays , Biomarkers, Tumor/genetics , Heterografts , Gene Expression Regulation, Neoplastic , Gene Expression Profiling
2.
Clin Cancer Res ; 26(18): 4933-4946, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32576626

ABSTRACT

PURPOSE: Advances in prostate cancer lag behind other tumor types partly due to the paucity of models reflecting key milestones in prostate cancer progression. Therefore, we develop clinically relevant prostate cancer models. EXPERIMENTAL DESIGN: Since 1996, we have generated clinically annotated patient-derived xenografts (PDXs; the MDA PCa PDX series) linked to specific phenotypes reflecting all aspects of clinical prostate cancer. RESULTS: We studied two cell line-derived xenografts and the first 80 PDXs derived from 47 human prostate cancer donors. Of these, 47 PDXs derived from 22 donors are working models and can be expanded either as cell lines (MDA PCa 2a and 2b) or PDXs. The histopathologic, genomic, and molecular characteristics (androgen receptor, ERG, and PTEN loss) maintain fidelity with the human tumor and correlate with published findings. PDX growth response to mouse castration and targeted therapy illustrate their clinical utility. Comparative genomic hybridization and sequencing show significant differences in oncogenic pathways in pairs of PDXs derived from different areas of the same tumor. We also identified a recurrent focal deletion in an area that includes the speckle-type POZ protein-like (SPOPL) gene in PDXs derived from seven human donors of 28 studied (25%). SPOPL is a SPOP paralog, and SPOP mutations define a molecular subclass of prostate cancer. SPOPL deletions are found in 7% of The Cancer Genome Atlas prostate cancers, which suggests that our cohort is a reliable platform for targeted drug development. CONCLUSIONS: The MDA PCa PDX series is a dynamic resource that captures the molecular landscape of prostate cancers progressing under novel treatments and enables optimization of prostate cancer-specific, marker-driven therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Precision Medicine/methods , Prostatic Neoplasms/drug therapy , Adaptor Proteins, Vesicular Transport/genetics , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Comparative Genomic Hybridization , DNA Copy Number Variations , Humans , Male , Mice , Primary Cell Culture , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Sequence Deletion , Xenograft Model Antitumor Assays/methods
3.
Endocr Relat Cancer ; 27(7): R255-R265, 2020 07.
Article in English | MEDLINE | ID: mdl-32369771

ABSTRACT

Many solid tumors metastasize to bone, but only prostate cancer has bone as a single, dominant metastatic site. Recently, the FGF axis has been implicated in cancer progression in some tumors and mounting evidence indicate that it mediates prostate cancer bone metastases. The FGF axis has an important role in bone biology and mediates cell-to-cell communication. Therefore, we discuss here basic concepts of bone biology, FGF signaling axis, and FGF axis function in adult bone, to integrate these concepts in our current understanding of the role of FGF axis in bone metastases.


Subject(s)
Bone Neoplasms/secondary , Fibroblast Growth Factors/metabolism , Female , Humans , Male , Signal Transduction
4.
Clin Cancer Res ; 23(17): 5135-5148, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28512172

ABSTRACT

Purpose: Conditioning strategies constitute a relatively unexplored and exciting opportunity to shape tumor fate by targeting the tumor microenvironment. In this study, we assessed how hemin, a pharmacologic inducer of heme oxygenase-1 (HO-1), has an impact on prostate cancer development in an in vivo conditioning model.Experimental Design: The stroma of C57BL/6 mice was conditioned by subcutaneous administration of hemin prior to TRAMP-C1 tumor challenge. Complementary in vitro and in vivo assays were performed to evaluate hemin effect on both angiogenesis and the immune response. To gain clinical insight, we used prostate cancer patient-derived samples in our studies to assess the expression of HO-1 and other relevant genes.Results: Conditioning resulted in increased tumor latency and decreased initial growth rate. Histologic analysis of tumors grown in conditioned mice revealed impaired vascularization. Hemin-treated human umbilical vein endothelial cells (HUVEC) exhibited decreased tubulogenesis in vitro only in the presence of TRAMP-C1-conditioned media. Subcutaneous hemin conditioning hindered tumor-associated neovascularization in an in vivo Matrigel plug assay. In addition, hemin boosted CD8+ T-cell proliferation and degranulation in vitro and antigen-specific cytotoxicity in vivo A significant systemic increase in CD8+ T-cell frequency was observed in preconditioned tumor-bearing mice. Tumors from hemin-conditioned mice showed reduced expression of galectin-1 (Gal-1), key modulator of tumor angiogenesis and immunity, evidencing persistent remodeling of the microenvironment. We also found a subset of prostate cancer patient-derived xenografts and prostate cancer patient samples with mild HO-1 and low Gal-1 expression levels.Conclusions: These results highlight a novel function of a human-used drug as a means of boosting the antitumor response. Clin Cancer Res; 23(17); 5135-48. ©2017 AACR.


Subject(s)
Galectin 1/genetics , Heme Oxygenase-1/genetics , Hemin/administration & dosage , Prostatic Neoplasms/drug therapy , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Disease Models, Animal , Galectin 1/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Heme Oxygenase-1/antagonists & inhibitors , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Male , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
5.
Cell Death Dis ; 7(12): e2570, 2016 12 29.
Article in English | MEDLINE | ID: mdl-28032857

ABSTRACT

Prostate cancer (PCa) cells display abnormal expression of cytoskeletal proteins resulting in an augmented capacity to resist chemotherapy and colonize distant organs. We have previously shown that heme oxygenase 1 (HO-1) is implicated in cell morphology regulation in PCa. Here, through a multi 'omics' approach we define the HO-1 interactome in PCa, identifying HO-1 molecular partners associated with the integrity of the cellular cytoskeleton. The bioinformatics screening for these cytoskeletal-related partners reveal that they are highly misregulated in prostate adenocarcinoma compared with normal prostate tissue. Under HO-1 induction, PCa cells present reduced frequency in migration events, trajectory and cell velocity and, a significant higher proportion of filopodia-like protrusions favoring zippering among neighboring cells. Moreover forced expression of HO-1 was also capable of altering cell protrusions in transwell co-culture systems of PCa cells with MC3T3 cells (pre-osteoblastic cell line). Accordingly, these effects were reversed under siHO. Transcriptomics profiling evidenced significant modulation of key markers related to cell adhesion and cell-cell communication under HO-1 induction. The integration from our omics-based research provides a four molecular pathway foundation (ANXA2/HMGA1/POU3F1; NFRSF13/GSN; TMOD3/RAI14/VWF; and PLAT/PLAU) behind HO-1 regulation of tumor cytoskeletal cell compartments. The complementary proteomics and transcriptomics approaches presented here promise to move us closer to unravel the molecular framework underpinning HO-1 involvement in the modulation of cytoskeleton pathways, pushing toward a less aggressive phenotype in PCa.


Subject(s)
Cell Communication/genetics , Gene Regulatory Networks , Heme Oxygenase-1/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Pseudopodia/metabolism , Animals , Cell Communication/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Coculture Techniques , Crystallography, X-Ray , Culture Media, Conditioned/pharmacology , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks/drug effects , Humans , Male , Mice , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/genetics , Protein Binding/drug effects , Proteomics , Pseudopodia/drug effects , Sequence Analysis, RNA , Tandem Mass Spectrometry , Transcriptome/drug effects , Transcriptome/genetics
6.
Oncotarget ; 7(14): 18798-811, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26933806

ABSTRACT

Metabolic syndrome (MeS) has been identified as a risk factor for breast cancer. C-terminal binding protein 1 (CtBP1) is a co-repressor of tumor suppressor genes that is activated by low NAD+/NADH ratio. High fat diet (HFD) increases intracellular NADH. We investigated the effect of CtBP1 hyperactivation by HFD intake on mouse breast carcinogenesis. We generated a MeS-like disease in female mice by chronically feeding animals with HFD. MeS increased postnatal mammary gland development and generated prominent duct patterns with markedly increased CtBP1 and Cyclin D1 expression. CtBP1 induced breast cancer cells proliferation. Serum from animals with MeS enriched the stem-like/progenitor cell population from breast cancer cells. CtBP1 increased breast tumor growth in MeS mice modulating multiple genes and miRNA expression implicated in cell proliferation, progenitor cells phenotype, epithelial to mesenchymal transition, mammary development and cell communication in the xenografts. These results define a novel function for CtBP1 in breast carcinogenesis.


Subject(s)
Alcohol Oxidoreductases/metabolism , Breast Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Metabolic Syndrome/metabolism , MicroRNAs/metabolism , Animals , Breast Neoplasms/genetics , Diet, High-Fat , Female , Heterografts , Humans , MCF-7 Cells , Metabolic Syndrome/genetics , Mice , Mice, Nude , NIH 3T3 Cells , Random Allocation , Risk Factors
7.
Sci Transl Med ; 6(252): 252ra122, 2014 Sep 03.
Article in English | MEDLINE | ID: mdl-25186177

ABSTRACT

Bone is the most common site of prostate cancer (PCa) progression to a therapy-resistant, lethal phenotype. We found that blockade of fibroblast growth factor receptors (FGFRs) with the receptor tyrosine kinase inhibitor dovitinib has clinical activity in a subset of men with castration-resistant PCa and bone metastases. Our integrated analyses suggest that FGF signaling mediates a positive feedback loop between PCa cells and bone cells and that blockade of FGFR1 in osteoblasts partially mediates the antitumor activity of dovitinib by improving bone quality and by blocking PCa cell-bone cell interaction. These findings account for clinical observations such as reductions in lesion size and intensity on bone scans, lymph node size, and tumor-specific symptoms without proportional declines in serum prostate-specific antigen concentration. Our findings suggest that targeting FGFR has therapeutic activity in advanced PCa and provide direction for the development of therapies with FGFR inhibitors.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Prostatic Neoplasms/pathology , Quinolones/therapeutic use , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Benzimidazoles/pharmacology , Bone Neoplasms/pathology , Bone and Bones/drug effects , Bone and Bones/metabolism , Cell Line, Tumor , Disease Models, Animal , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Osteoblasts/drug effects , Osteoblasts/metabolism , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Quinolones/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Stromal Cells/drug effects , Stromal Cells/pathology , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
8.
Clin Cancer Res ; 20(15): 4086-95, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24842953

ABSTRACT

PURPOSE: Clinical and epidemiologic data suggest that obesity is associated with more aggressive forms of prostate cancer, poor prognosis, and increased mortality. C-terminal-binding protein 1 (CtBP1) is a transcription repressor of tumor suppressor genes and is activated by NADH binding. High calorie intake decreases intracellular NAD(+)/NADH ratio. The aim of this work was to assess the effect of high-fat diet (HFD) and CtBP1 expression modulation over prostate xenograft growth. EXPERIMENTAL DESIGN: We developed a metabolic syndrome-like disease in vivo model by feeding male nude mice with HFD during 16 weeks. Control diet (CD)-fed animals were maintained at the same conditions. Mice were inoculated with PC3 cells stable transfected with shCtBP1 or control plasmids. Genome-wide expression profiles and Gene Set Enrichment Analysis (GSEA) were performed from PC3.shCtBP1 versus PC3.pGIPZ HFD-fed mice tumors. RESULTS: No significant differences were observed in tumor growth on CD-fed mice; however, we found that only 60% of HFD-fed mice inoculated with CtBP1-depleted cells developed a tumor. Moreover these tumors were significantly smaller than those generated by PC3.pGIPZ control xenografts. We found 823 genes differentially expressed in shCtBP1 tumors from HFD-fed mice. GSEA from expression dataset showed that most of these genes correspond to cell adhesion, metabolic process, and cell cycle. CONCLUSIONS: Metabolic syndrome-like diseases and CtBP1 expression cooperate to induce prostate tumor growth. Hence, targeting of CtBP1 expression might be considered for prostate cancer management and therapy in the subset of patients with metabolic syndromes.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , DNA-Binding Proteins/antagonists & inhibitors , Diet, High-Fat/adverse effects , Metabolic Syndrome/prevention & control , Obesity/prevention & control , Prostatic Neoplasms/prevention & control , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Adhesion , Cell Proliferation , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Gonadal Steroid Hormones/pharmacology , Humans , Immunoenzyme Techniques , Male , Metabolic Syndrome/etiology , Metabolic Syndrome/pathology , Mice , Mice, Nude , Obesity/etiology , Obesity/pathology , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Clin Cancer Res ; 18(3): 726-36, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22298898

ABSTRACT

PURPOSE: To study Wnt/ß-catenin in castrate-resistant prostate cancer (CRPC) and understand its function independently of the ß-catenin-androgen receptor (AR) interaction. EXPERIMENTAL DESIGN: We carried out ß-catenin immunocytochemical analysis, evaluated TOP-flash reporter activity (a reporter of ß-catenin-mediated transcription), and sequenced the ß-catenin gene in MDA prostate cancer 118a, MDA prostate cancer 118b, MDA prostate cancer 2b, and PC-3 prostate cancer cells. We knocked down ß-catenin in AR-negative MDA prostate cancer 118b cells and carried out comparative gene-array analysis. We also immunohistochemically analyzed ß-catenin and AR in 27 bone metastases of human CRPCs. RESULTS: ß-Catenin nuclear accumulation and TOP-flash reporter activity were high in MDA prostate cancer 118b but not in MDA prostate cancer 2b or PC-3 cells. MDA prostate cancer 118a and MDA prostate cancer 118b cells carry a mutated ß-catenin at codon 32 (D32G). Ten genes were expressed differently (false discovery rate, 0.05) in MDA prostate cancer 118b cells with downregulated ß-catenin. One such gene, hyaluronan synthase 2 (HAS2), synthesizes hyaluronan, a core component of the extracellular matrix. We confirmed HAS2 upregulation in PC-3 cells transfected with D32G-mutant ß-catenin. Finally, we found nuclear localization of ß-catenin in 10 of 27 human tissue specimens; this localization was inversely associated with AR expression (P = 0.056, Fisher's exact test), suggesting that reduced AR expression enables Wnt/ß-catenin signaling. CONCLUSION: We identified a previously unknown downstream target of ß-catenin, HAS2, in prostate cancer, and found that high ß-catenin nuclear localization and low or no AR expression may define a subpopulation of men with bone metastatic prostate cancer. These findings may guide physicians in managing these patients.


Subject(s)
Glucuronosyltransferase/genetics , Prostatic Neoplasms/genetics , Signal Transduction/physiology , beta Catenin/genetics , Animals , Blotting, Western , Bone Neoplasms/secondary , Gene Expression Profiling , Glucuronosyltransferase/metabolism , Humans , Hyaluronan Synthases , Immunohistochemistry , Male , Mice , Mice, SCID , Mutation , Prostatic Neoplasms/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , beta Catenin/metabolism
10.
Mol Cancer Res ; 9(8): 1078-90, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21700680

ABSTRACT

BRCA1 plays numerous roles in the regulation of genome integrity and chemoresistance. Although BRCA1 interaction with key proteins involved in DNA repair is well known, its role as a coregulator in the transcriptional response to DNA damage remains poorly understood. In this study, we show that BRCA1 plays a central role in the transcriptional response to genotoxic stress in prostate cancer. BRCA1 expression mediates apoptosis, cell-cycle arrest, and decreased viability in response to doxorubicin treatment. Xenograft studies using human prostate carcinoma PC3 cells show that BRCA1 depletion results in increased tumor growth. A focused survey of BRCA1-regulated genes in prostate carcinoma reveals that multiple regulators of genome stability and cell-cycle control, including BLM, FEN1, DDB2, H3F3B, BRCA2, CCNB2, MAD2L1, and GADD153, are direct transcriptional targets of BRCA1. Furthermore, we show that BRCA1 targets GADD153 promoter to increase its transcription in response to DNA damage. Finally, GADD153 depletion significantly abrogates BRCA1 influence on cell-cycle progression and cell death in response to doxorubicin treatment. These findings define a novel transcriptional pathway through which BRCA1 orchestrates cell fate decisions in response to genotoxic insults, and suggest that BRCA1 status should be considered for new chemotherapeutic treatment strategies in prostate cancer.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , BRCA1 Protein/metabolism , Carcinoma/metabolism , DNA Damage/genetics , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Prostatic Neoplasms/metabolism , Transcription Factor CHOP/metabolism , Animals , BRCA1 Protein/genetics , Carcinoma/pathology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Genomic Instability/genetics , Humans , Male , Mice , Prostatic Neoplasms/pathology , Transcription Factor CHOP/genetics
11.
Mol Cancer Res ; 7(11): 1745-55, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19903769

ABSTRACT

Prostate cancer (PCa) is the second leading cause of cancer-associated death in men. Inflammation has been recognized as a risk factor for this disease. Heme oxygenase 1 (HO-1), the inducible isoform of the rate-limiting enzyme in heme degradation, counteracts oxidative and inflammatory damage. Here, we investigated the regulated expression of HO-1 and its functional consequences in PCa. We studied the effect of genetic and pharmacologic disruption of HO-1 in the growth, invasion, and migration in androgen-sensitive (MDA PCa2b and LNCaP) and androgen-insensitive (PC3) PCa cell lines. Our results show that HO-1 levels are markedly decreased in PC3 compared with MDA PCa2b and LNCaP. Hemin treatment increased HO-1 at both protein and mRNA levels in all cell lines and decreased cell proliferation and invasion. Furthermore, overexpression of HO-1 in PC3 resulted in markedly reduced cell proliferation and migration. Accordingly, small interfering RNA-mediated silencing of HO-1 expression in MDA PCa2b cells resulted in increased proliferation and invasion. Using reverse transcription-quantitative PCR-generated gene array, a set of inflammatory and angiogenic genes were upregulated or downregulated in response to HO-1 overexpression identifying matrix metalloprotease 9 (MMP9) as a novel downstream target of HO-1. MMP9 production and activity was downregulated by HO-1 overexpression. Furthermore, PC3 cells stably transfected with HO-1 (PC3HO-1) and controls were injected into nu/nu mice for analysis of in vivo tumor xenograft phenotype. Tumor growth and MMP9 expression was significantly reduced in PC3HO-1 tumors compared with control xenografts. Taken together, these results implicate HO-1 in PCa cell migration and proliferation suggesting its potential role as a therapeutic target in clinical settings.


Subject(s)
Heme Oxygenase-1/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Animals , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Down-Regulation , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Heme Oxygenase-1/genetics , Hemin/pharmacology , Humans , Immunohistochemistry , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Microarray Analysis , Neoplasm Invasiveness , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Transfection , Transplantation, Heterologous
12.
J Clin Invest ; 118(8): 2697-710, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18618013

ABSTRACT

In prostate cancer, androgen blockade strategies are commonly used to treat osteoblastic bone metastases. However, responses to these therapies are typically brief, and the mechanism underlying androgen-independent progression is not clear. Here, we established what we believe to be the first human androgen receptor-negative prostate cancer xenografts whose cells induced an osteoblastic reaction in bone and in the subcutis of immunodeficient mice. Accordingly, these cells grew in castrated as well as intact male mice. We identified FGF9 as being overexpressed in the xenografts relative to other bone-derived prostate cancer cells and discovered that FGF9 induced osteoblast proliferation and new bone formation in a bone organ assay. Mice treated with FGF9-neutralizing antibody developed smaller bone tumors and reduced bone formation. Finally, we found positive FGF9 immunostaining in prostate cancer cells in 24 of 56 primary tumors derived from human organ-confined prostate cancer and in 25 of 25 bone metastasis cases studied. Collectively, these results suggest that FGF9 contributes to prostate cancer-induced new bone formation and may participate in the osteoblastic progression of prostate cancer in bone. Androgen receptor-null cells may contribute to the castration-resistant osteoblastic progression of prostate cancer cells in bone and provide a preclinical model for studying therapies that target these cells.


Subject(s)
Bone Neoplasms/secondary , Fibroblast Growth Factor 9/metabolism , Osteogenesis/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Karyotyping , Keratins/metabolism , Male , Mice , Mice, SCID , Middle Aged , Neoplasm Metastasis , Neoplasm Transplantation , Orchiectomy , Organ Culture Techniques , Osteoblasts/metabolism , Prostatic Neoplasms/genetics , Transplantation, Heterologous , Vimentin/metabolism
13.
BMC Cancer ; 6: 286, 2006 Dec 14.
Article in English | MEDLINE | ID: mdl-17169158

ABSTRACT

BACKGROUND: Chronic injury deregulates cellular homeostasis and induces a number of alterations leading to disruption of cellular processes such as cell cycle checkpoints and apoptosis, driving to carcinogenesis. The stress protein heme oxygenase-1 (HO-1) catalyzes heme degradation producing biliverdin, iron and CO. Induction of HO-1 has been suggested to be essential for a controlled cell growth. The aim of this work was to analyze the in vivo homeostatic response (HR) triggered by the withdrawal of a potent carcinogen, p-dimethylaminoazobenzene (DAB), after preneoplastic lesions were observed. We analyzed HO-1 cellular localization and the expression of HO-1, Bcl-2 and cell cycle related proteins under these conditions comparing them to hepatocellular carcinoma (HC). METHODS: The intoxication protocol was designed based on previous studies demonstrating that preneoplastic lesions were evident after 89 days of chemical carcinogen administration. Male CF1 mice (n = 18) were used. HR group received DAB (0.5 % w/w) in the diet for 78 days followed by 11 days of carcinogen deprivation. The HC group received the carcinogen and control animals the standard diet during 89 days. The expression of cell cycle related proteins, of Bcl-2 and of HO-1 were analyzed by western blot. The cellular localization and expression of HO-1 were detected by immnunohistochemistry. RESULTS: Increased expression of cyclin E/CDK2 was observed in HR, thus implicating cyclin E/CDK2 in the liver regenerative process. p21cip1/waf1 and Bcl-2 induction in HC was restituted to basal levels in HR. A similar response profile was found for HO-1 expression levels, showing a lower oxidative status in the carcinogen-deprived liver. The immunohistochemical studies revealed the presence of macrophages surrounding foci of necrosis and nodular lesions in HR indicative of an inflammatory response. Furthermore, regenerative cells displayed changes in type, size and intensity of HO-1 immunostaining. CONCLUSION: These results demonstrate that the regenerative capacity of the liver is still observed in the pre-neoplastic tissue after carcinogen withdrawal suggesting that reversible mechanism/s to compensate necrosis and to restitute homeostasis are involved.


Subject(s)
Carcinogens/toxicity , Cell Cycle Proteins/genetics , Cell Cycle/drug effects , Heme Oxygenase-1/genetics , p-Dimethylaminoazobenzene/toxicity , Animals , Carcinogens/administration & dosage , Carcinoma, Hepatocellular/enzymology , Drug Administration Schedule , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Homeostasis , Liver Neoplasms/enzymology , Male , Mice , Mice, Inbred Strains , p-Dimethylaminoazobenzene/administration & dosage
14.
Per Med ; 1(1): 127-130, 2004 Dec.
Article in English | MEDLINE | ID: mdl-29793225

ABSTRACT

Genomics must be combined with proteomics and metabolomics to rationalize a therapeutic strategy that considers gene expression, protein expression and metabolic profiles in the target organ to gain insight into other pathways implicated in the same or contributory tissues. Multidisciplinary strategies such as this provide an interactive process by which findings are translated into novel therapies.

15.
Carcinogenesis ; 23(8): 1289-96, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12151346

ABSTRACT

Current therapy for advanced prostate cancer is largely based on androgen deprivation and is mostly palliative because all patients eventually relapse with androgen-independent disease. Doxorubicin (Dx), an anthracycline used commonly as a chemotherapeutic agent in relapsed prostate cancer, is a strong inducer of p53 expression and p21(CIP1/WAF1) (p21) transactivation. Previous reports suggest that p21 may have a role in the modulation to chemotherapy-induced apoptosis, prostate cancer progression and androgen regulation. In order to investigate if p21 has a pro-survival role in the response of prostate cancer cells to cellular stress, we exposed two androgen-regulated human prostate cancer cell lines (MDA PCa 2b and LNCaP) to Dx and growth factor withdrawal. We then studied expression of p53 and p21, cell-cycle kinetics and apoptosis. We have found that p53 protein accumulated in a dose- and time-dependent manner after Dx treatment, while p21 expression increased over time with low but decreased with high Dx doses. Apoptosis occurred in parallel with p21 down-modulation. Dx treatment of p53 knockout cells demonstrated that p21 induction was strictly p53 dependent. Reduction of p21 levels in prostate cancer cells with an antisense p21 adenovirus resulted in sensitization to Dx and accelerated onset of apoptosis in response to growth factor withdrawal. The evidence presented here also suggests that caspase activation mediates the apoptosis in this system and supports that p21 may modulate the threshold of apoptosis in prostate cancer. These observations may thus provide implications onto the integration of chemotherapy and androgen ablation.


Subject(s)
Apoptosis/physiology , Cyclins/physiology , DNA Damage , Growth Substances/metabolism , Neoplasms, Hormone-Dependent/pathology , Prostatic Neoplasms/pathology , Antibiotics, Antineoplastic/pharmacology , Cell Cycle , Cell Survival , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Doxorubicin/pharmacology , Humans , Male , Neoplasms, Hormone-Dependent/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/physiology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
16.
BMC Cancer ; 2: 6, 2002 Mar 22.
Article in English | MEDLINE | ID: mdl-11914144

ABSTRACT

BACKGROUND: Acute Intermittent Porphyria is a genetic disorder of heme metabolism, characterized by increased levels of porphyrin precursors, delta-aminolevulinic acid (ALA) and porphobilinogen (PBG). ALA has been reported to generate reactive oxygen species and to cause oxidative damage to proteins, subcellular structures and DNA. It is known that oxidative stress can induce apoptosis. The aim of this work was to study the cytotoxic effect of ALA on two hepatocarcinoma cell lines. RESULTS: We have determined the impact of ALA on HEP G2 and HEP 3B hepatocarcinoma cell lines survival as measured by the MTT assay. ALA proved to be cytotoxic in both cell lines however; HEP G2 was more sensitive to ALA than HEP 3B. Addition of hemin or glucose diminished ALA cytotoxicity in HEP G2 cells; instead it was enhanced in HEP 3B cells. Because apoptosis is usually associated with DNA fragmentation, the DNA of ALA treated and untreated cells were analyzed. The characteristic pattern of DNA fragmentation ladders was observed in ALA treated cells. To elucidate the mechanisms of ALA induced apoptosis, we examined its effect on p53 expression. No changes in p53 mRNA levels were observed after exposure of both cell lines to ALA for 24 h. CDK2 and CDK4 protein levels were reduced after ALA treatment at physiological concentrations.


Subject(s)
Aminolevulinic Acid/toxicity , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Aminolevulinic Acid/therapeutic use , Carcinoma, Hepatocellular/metabolism , Cell Cycle/drug effects , DNA Fragmentation/drug effects , DNA, Neoplasm/metabolism , Dose-Response Relationship, Drug , Glucose/pharmacology , Hemin/pharmacology , Humans , Liver Neoplasms/metabolism , Tetrazolium Salts/metabolism , Thiazoles/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...