Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 116(44): 22246-22251, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31611368

ABSTRACT

Immune checkpoint (IC) therapy provides substantial benefits to cancer patients but can also cause distinctive toxicities termed immune-related adverse events (irAEs). Biomarkers to predict toxicities will be necessary to improve management of patients receiving IC therapy. We relied on serological analysis of recombinant cDNA expression libraries to evaluate plasma samples from patients treated with IC therapy and identified autoantibodies, both in pretreatment and on-treatment samples prior to the development of irAEs, which correlate with the development of immune-related hypophysitis (anti-GNAL and anti-ITM2B autoantibodies) and pneumonitis (anti-CD74 autoantibody). We developed an enzyme-linked immunosorbent assay and tested additional patient samples to confirm our initial findings. Collectively, our data suggest that autoantibodies may correlate with irAEs related to IC therapy, and specific autoantibodies may be detected early for the management of irAEs.


Subject(s)
Autoantibodies/immunology , Autoimmune Hypophysitis/etiology , Immunotherapy/adverse effects , Pneumonia/etiology , Adaptor Proteins, Signal Transducing/immunology , Aged , Autoantibodies/blood , Autoantibodies/toxicity , Autoimmune Hypophysitis/diagnosis , Autoimmune Hypophysitis/immunology , Biomarkers/blood , Female , GTP-Binding Protein alpha Subunits, Gi-Go/immunology , Humans , Male , Middle Aged , Neoplasms/therapy , Pneumonia/immunology
2.
Proc Natl Acad Sci U S A ; 113(42): 11919-11924, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27698113

ABSTRACT

Immune checkpoint therapies, such as ipilimumab, induce dramatic antitumor responses in a subset of patients with advanced malignancies, but they may also induce inflammatory responses and toxicities termed immune-related adverse events (irAEs). These irAEs are often low grade and manageable, but severe irAEs may lead to prolonged hospitalizations or fatalities. Early intervention is necessary to minimize morbidities that occur with severe irAEs. However, correlative biomarkers are currently lacking. In a phase II clinical trial that treated 27 patients with metastatic prostate cancer, we aimed to test the safety and efficacy of androgen deprivation therapy plus ipilimumab. In this study, we observed grade 3 toxicities in >40% of treated patients, which led to early closure of the study. Because ipilimumab enhances T-cell responses, we hypothesized that increased clonal T-cell responses in the systemic circulation may contribute to irAEs. Sequencing of the T-cell receptor ß-chains in purified T cells revealed clonal expansion of CD8 T cells, which occurred in blood samples collected before the onset of grade 2-3 irAEs. These initial results suggested that expansion of ≥55 CD8 T-cell clones preceded the development of severe irAEs. We further evaluated available blood samples from a second trial and determined that patients who experienced grade 2-3 irAEs also had expansion of ≥55 CD8 T-cell clones in blood samples collected before the onset of irAEs. We propose that CD8 T-cell clonal expansion may be a correlative biomarker to enable close monitoring and early intervention for patients receiving ipilimumab.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , CD8-Positive T-Lymphocytes/immunology , Clonal Evolution/immunology , Drug-Related Side Effects and Adverse Reactions/etiology , Ipilimumab/adverse effects , Lymphocyte Count , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers , CD8-Positive T-Lymphocytes/metabolism , Clinical Trials, Phase II as Topic , Disease Susceptibility , Drug-Related Side Effects and Adverse Reactions/diagnosis , Humans , Ipilimumab/therapeutic use , Male , Middle Aged , Prostatic Neoplasms/complications , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Severity of Illness Index , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Treatment Outcome
3.
Cancer Res ; 73(6): 1900-11, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23302227

ABSTRACT

Caveolin 1 (Cav-1) is a plasma membrane-associated protein with the capacity to modulate signaling activities in a context-dependent fashion. Interactions between Cav-1 and low-density lipoprotein receptor-related protein 6 (LRP6) were reported to be important for the regulation of Wnt-ß-catenin (ß-cat) signaling. Cav-1 also interacts with insulin and IGF-I receptors (IGF-IR/IR) and can stimulate IR kinase activities. We found positive correlation between Cav-1 and LRP6 expression in both human primary prostate cancer and metastasis tissues and in PC-3 cells. Cav-1 stimulation of Wnt-ß-cat signaling and c-Myc levels was positively associated with LRP6 expression in LNCaP, PC-3, and DU145 prostate cancer cells. Importantly, LRP6 and, to a lesser extent, Cav-1 were found to stimulate aerobic glycolysis. These activities were positively associated with the expression of HK2 and Glut3 and shown to be dependent on Akt signaling by both gene knockdown and chemical inhibition methods. We further showed that Cav-1 and LRP6 exert their effects on Akt and glycolytic activities by stimulating IGF-IR/IR signaling. Overall, our results show that Cav-1 interacts with LRP6 to generate an integrated signaling module that leads to the activation of IGF-IR/IR and results in stimulation of Akt-mTORC1 signaling and aerobic glycolysis in prostate cancer.


Subject(s)
Caveolin 1/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Prostatic Neoplasms/metabolism , Signal Transduction , Aerobiosis , Cell Line, Tumor , Glycolysis , Humans , Immunohistochemistry , Male , Phosphorylation , Prostatic Neoplasms/pathology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
4.
Cancer Biol Ther ; 14(2): 117-26, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23114714

ABSTRACT

We investigated the effect of dasatinib and sunitinib on tyrosine kinase (TK) signaling, caveolin-1 (Cav-1) expression and secretion and proliferation of PC-3 and DU145 prostate cancer cells in vitro and in vivo. Treatment of both cell lines with either dasatinib or sunitinib reduced phosphorylation of PDGFR, VEGFR2, Akt, FAK, Src (dasatinib only) and Cav-1, and reduced cellular and secreted levels of Cav-1. Both agents dose-dependently inhibited proliferation of these cells. In PC-3 and DU145 subcutaneous xenografts, treatment with dasatinib, sunitinib or anti-Cav-1 antibody (Ab) alone produced significant tumor regression compared with that by vehicle or IgG alone. Combined dasatinib and anti-Cav-1 Ab treatment or sunitinib and anti-Cav-1 Ab produced greater tumor regression than either treatment alone. Serum Cav-1 levels were lower in dasatinib- and sunitinib-treated mice than they were in vehicle-treated mice, and correlated positively with tumor growth in dasatinib- and sunitinib-treated groups (r = 0.48, p = 0.031; r = 0.554, p = 0.0065, respectively), compared with vehicle controls. Cav-1 knockdown, in combination with dasatinib or sunitinib treatment in PC-3 cells, caused a greater reduction in the phosphorylation of PDGFR-ß and VEGFR2, and expression and secretion of PDGF-B and VEGF-A than that in PC-3 cells treated with dasatinib or sunitinib alone in control siRNA cells, suggesting that Cav-1 is involved in an autocrine pathway that is affected by these drugs. Overall, our results suggest a role for Cav-1 as a biomarker of response to both dasatinib and sunitinib treatment and as a therapeutic target in prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor , Caveolin 1/blood , Prostatic Neoplasms/blood , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Humans , Male , Mice , Prostatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Treatment Outcome , Xenograft Model Antitumor Assays
5.
Hybridoma (Larchmt) ; 31(2): 77-86, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22509911

ABSTRACT

Caveolae are critical cell surface structures important in coordinated cell signaling and endocytosis. One of the major proteins of caveolae is caveolin 1 (Cav-1). Cellular levels of Cav-1 are associated with cancer progression. In prostate cancer cells, levels of Cav-1 are positively correlated with tumor progression and metastasis. Cav-1 can be secreted by prostate cancer cells into the microenvironment and triggers proliferation and anti-apoptosis of the tumor and tumor endothelial cells. Clinical studies have shown increased serum Cav-1 levels in patients with poor prognosis. In tissue culture and animal model experiments, blocking secreted Cav-1 by polyclonal antibodies inhibits tumor cell growth. Cav-1 is therefore a potential therapeutic target for prostate cancer treatment. In this study, we used Cav-1 knock-out mice as hosts to produce monoclonal anti-Cav-1 antibodies. A total of 11 hybridoma cell lines were selected for their ability to produce antibodies that bound GST-Cav-1 but not GST on glutathione-coated ELISA plates. Further screening with ELISAs using GST-Cav-1 fragments on GSH-coated plates classified these antibodies into four groups: N1-31 with five antibodies binds the far N-terminus between amino acids 1 and 31; N32-80 with three antibodies binds between amino acids 32 and 80; CSD with two antibodies potentially bind the scaffolding domain (amino acids 80-101); and Cav-1-C with 1 antibody binds parts of the C-terminal half. Binding affinities (Kd) of these antibodies to soluble Cav-1 ranged from 10(-11) to 10(-8) M. Binding competition experiments revealed that these antibodies recognized a total of six different epitopes on Cav-1. Potency of these antibodies to neutralize Cav-1-mediated signaling pathways in cultured cells and in animal models will be tested. A selected monoclonal antibody will then be humanized and be further developed into a potential anti-prostate cancer therapeutic.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Caveolin 1/immunology , Prostatic Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/isolation & purification , Antibody Specificity , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Line, Tumor , Humans , Hybridomas/immunology , Male , Mice , Mice, Knockout , Prostatic Neoplasms/pathology , Recombinant Proteins/genetics , Recombinant Proteins/immunology
7.
Prostate ; 70(9): 1020-35, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20209490

ABSTRACT

BACKGROUND: Caveolin-1 (cav-1) is overexpressed by metastatic prostate cancer (PC) cells. Pre-operative serum cav-1 levels have been shown to be a prognostic marker for PC recurrence. This study evaluated the relationship between post-treatment serum cav-1 levels and single nucleotide polymorphisms (SNPs) in the cav-1 and -2 genes with risk of PC, aggressive PC, PC recurrence or death. METHODS: Two case-control studies of PC among men in Washington State were combined for this analysis. Cases (n = 1,458) were diagnosed in 1993-1996 or 2002-2005 and identified via a SEER cancer registry. Age-matched controls (n = 1,351) were identified via random digit dialing. Logistic regression was used to assess the relationship between exposures (19 haplotype-tagging SNPs from all subjects and post-treatment serum cav-1 levels from a sample of 202 cases and 226 controls) and PC risk and aggressive PC. Cox proportional hazards regression was used to assess the relationship between exposures and PC recurrence and death. RESULTS: Rs9920 in cav-1 was associated with an increased relative risk of overall PC (OR(CT + CC) = 1.37, 95% CI = 1.12, 1.68) and aggressive PC (OR(CT + CC) = 1.57, 95% CI = 1.20, 2.06), but not with PC recurrence or death. High post-treatment serum cav-1 levels were not associated with PC risk, aggressive PC, or PC-specific death, but approached a significant inverse association with PC recurrence (hazard ratio = 0.69, 95% CI = 0.47, 1.00). CONCLUSIONS: We found modest evidence for an association with a variant in the cav-1 gene and risk of overall PC and aggressive PC, which merits further study. We found no evidence that higher post-treatment serum cav-1 is associated with risk of aggressive PC or adverse PC outcomes.


Subject(s)
Caveolin 1/blood , Caveolin 1/genetics , Caveolin 2/genetics , Prostatic Neoplasms/genetics , Adult , Aged , Alleles , Biomarkers, Tumor/genetics , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Variation , Haplotypes , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Polymorphism, Single Nucleotide/genetics , Proportional Hazards Models , Prostatic Neoplasms/blood , Prostatic Neoplasms/therapy , Regression Analysis , Risk Assessment , SEER Program , Treatment Outcome
8.
Cancer Biol Ther ; 8(23): 2286-96, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19923922

ABSTRACT

Caveolin-1 (cav-1) is a multifunctional protein and major component of caveolae membranes serving important functions related to signal transduction, endocytosis, transcytosis, and molecular transport. We previously showed that cav-1 is overexpressed and secreted by metastatic prostate cancer cells. We now report that cav-1 gene transduction (Adcav-1) or recombinant cav-1 (rcav-1) protein treatment of cav-1-negative prostate cancer cell line LP-LNCaP or cav-1(-/-) endothelial cells potentiated VEGF-stimulated angiogenic signaling. Downregulation of cav-1 in prostate cancer cell line PC-3 or human umbilical vein endothelial cells (HUVECs) through cav-1 siRNA significantly reduced basal and VEGF-stimulated phosphorylation of VEGFR2 (Y951), PLCgamma1 (Y783) and/or Akt (S473 & T308) relative to those in control siRNA treated cells. Additionally rcav-1 stimulation of cav-1 siRNA treated HUVECs restored this signaling pathway. Confocal microscopy and immunoprecipitation analysis revealed association and colocalization of VEGFR2 and PLCgamma1 with cav-1 following VEGF stimulation in HUVECs. Interestingly, treatment of HUVECs with cav-1 scaffolding domain (CSD) caused significant reduction in the VEGF-stimulated phosphorylation of VEGFR2, PLCgamma1 and Akt suggesting that CSD inhibits cav-1-mediated angiogenic signaling. VEGF stimulation of HUVECs significantly increased tubule length and cell migration, but this stimulatory effect was significantly reduced by cav-1 siRNA and/or CSD treatment. The present study demonstrates that cav-1 regulates VEGF-stimulated VEGFR2 autophosphorylation and activation of downstream angiogenic signaling, possibly through compartmentalization of specific signaling molecules. Our results provide mechanistic insight into the role of cav-1 in prostate cancer and suggest the use of CSD as a therapeutic tool to suppress angiogenic signaling in prostate cancer.


Subject(s)
Caveolin 1/physiology , Endothelium, Vascular/metabolism , Neovascularization, Physiologic , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/pathology , Umbilical Veins/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Aorta/cytology , Aorta/metabolism , Blotting, Western , Caveolin 1/antagonists & inhibitors , Cell Adhesion , Cell Movement , Cell Proliferation , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Knockout , Phospholipase C gamma/metabolism , Phosphorylation , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/pharmacology , Signal Transduction , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Wound Healing
9.
Mol Cancer Res ; 7(9): 1446-55, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19737975

ABSTRACT

Previously, we reported that caveolin-1 (cav-1) is overexpressed in metastatic prostate cancer and that virulent prostate cancer cells secrete biologically active cav-1. We also showed that cav-1 expression leads to prosurvival activities through maintenance of activated Akt and that cav-1 is taken up by other cav-1-negative tumor cells and/or endothelial cells, leading to stimulation of angiogenic activities through PI-3-K-Akt-eNOS signaling. To analyze the functional consequences of cav-1 overexpression on the development and progression of prostate cancer in vivo, we generated PBcav-1 transgenic mice. Adult male PBcav-1 mice showed significantly increased prostatic wet weight and higher incidence of epithelial hyperplasia compared with nontransgenic littermates. Increased immunostaining for cav-1, proliferative cell nuclear antigen, P-Akt, and reduced nuclear p27(Kip1) staining occurred in PBcav-1 hyperplastic prostatic lesions. PBcav-1 mice showed increased resistance to castration-induced prostatic regression and elevated serum cav-1 levels compared with nontransgenic littermates. Intraprostatic injection of androgen-sensitive, cav-1-secreting RM-9 mouse prostate cancer cells resulted in tumors that were larger in PBcav-1 mice than in nontransgenic littermates (P = 0.04). Tail vein inoculation of RM-9 cells produced significantly more experimental lung metastases in PBcav-1 males than in nontransgenic male littermates (P = 0.001), and in cav-1(+/+) mice than in cav-1(-/-) mice (P = 0.041). Combination treatment with surgical castration and systemic cav-1 antibody dramatically reduced the number of experimental metastases. These experimental data suggest a causal association of secreted cav-1 and prostate cancer growth and progression.


Subject(s)
Caveolin 1/metabolism , Prostatic Neoplasms/metabolism , Analysis of Variance , Androgen-Binding Protein/genetics , Androgens/metabolism , Animals , Caveolin 1/blood , Caveolin 1/genetics , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Lung Neoplasms/metabolism , Male , Mice , Mice, Transgenic , Neoplasm Metastasis , Neoplasm Transplantation , Orchiectomy , Promoter Regions, Genetic/genetics , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/blood , Proto-Oncogene Proteins c-akt/metabolism , Vascular Endothelial Growth Factor A/metabolism
10.
Cancer Res ; 68(3): 731-9, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18245473

ABSTRACT

Caveolin, a major structural component of specialized plasma membrane invaginations (caveolae) that participate in diverse cellular activities, has been implicated in the pathogenesis of several human diseases, including cancer. We showed in earlier studies that caveolin-1 (cav-1) is consistently and strongly overexpressed in metastatic prostate cancer and is secreted in a biologically active form by virulent prostate cancer cells. Using both in vitro and in vivo model systems, we now present evidence supporting a proangiogenic role for cav-1 in prostate cancer development and progression. Recombinant cav-1 (rcav-1) was taken up by cav-1(-/-) endothelial cells through either a lipid raft/caveolae- or clathrin-dependent mechanism, leading to specific angiogenic activities (tubule formation, cell migration, and nitric oxide production) that were mediated by rcav-1 stimulation of the PI3K-Akt-eNOS signaling module. Pathologic angiogenesis induced by cav-1 in prostate cancer-bearing mice correlated with an increased frequency, number, and size of lung metastases. We propose that in addition to its antiapoptotic role, cav-1 secreted by prostate cancer cells functions critically as a proangiogenic factor in metastatic progression of this tumor. These new insights into cav-1 function in prostate cancer may provide a base for the design of clinically applicable therapeutic strategies.


Subject(s)
Caveolin 1/physiology , Prostatic Neoplasms/blood supply , Animals , Caveolae/metabolism , Caveolin 1/metabolism , Caveolin 1/pharmacokinetics , Caveolin 1/pharmacology , Clathrin/metabolism , Endothelial Cells/metabolism , Humans , Male , Membrane Microdomains/metabolism , Mice , Mice, Nude , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Transfection
11.
Clin Cancer Res ; 12(16): 4872-5, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16914574

ABSTRACT

PURPOSE: Up-regulation of caveolin-1 (cav-1) is associated with virulent prostate cancer, and serum cav-1 levels are elevated in prostate cancer patients but not in benign prostatic hyperplasia. In this study, we evaluated the potential of high preoperative serum cav-1 levels to predict biochemical progression of prostate cancer. The value of the combined preoperative markers, prostate-specific antigen (PSA), biopsy Gleason score, and serum cav-1 for predicting biochemical recurrence was also investigated. EXPERIMENTAL DESIGN: Serum samples taken from 419 prostate cancer patients before radical prostatectomy were selected from our Specialized Programs of Research Excellence prostate cancer serum and tissue bank. Serum samples were obtained 0 to 180 days before surgery and all patients had complete data on age, sex, race, stage at enrollment, and follow-up for biochemical recurrence. Serum cav-1 levels were measured according to our previously reported ELISA protocol. RESULTS: Cav-1 levels were measured in the sera of 419 prostate cancer patients; the mean serum level was 4.52 ng/mL (median 1.01 ng/mL). Patients with high serum cav-1 levels had a 2.7-fold (P = 0.0493) greater risk of developing biochemical recurrence compared with those with low serum cav-1 levels. Importantly, patients with serum PSA >/= 10 ng/mL and elevated levels of serum cav-1 had 2.44 times higher risk (P = 0.0256) of developing biochemical recurrence compared with patients with low levels of cav-1. In addition, high serum cav-1 levels combined with increasing biopsy Gleason score predicted much shorter recurrence-free survival in the group of patients with PSA >/= 10 ng/mL (P = 0.0353). Cav-1 was also able to distinguish between high- and low- risk patients with biopsy Gleason score of seven, after adjusting, for patients PSA levels (P = 0.0429). CONCLUSIONS: Overall, elevated preoperative levels of serum cav-1 predict decreased time to cancer recurrence. In the subset of patients with serum PSA of >/=10 ng/mL, the combination of serum cav-1 and biopsy Gleason score has the capacity to predict time to biochemical recurrence.


Subject(s)
Biomarkers, Tumor/blood , Caveolin 1/blood , Neoplasm Recurrence, Local/blood , Prostatic Neoplasms/blood , Adult , Aged , Cohort Studies , Humans , Male , Middle Aged , Prognosis , Prostate-Specific Antigen/blood , Prostatectomy , Prostatic Neoplasms/surgery
12.
Cancer Res ; 66(10): 5159-64, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16707439

ABSTRACT

Caveolin-1 (cav-1) is a major scaffolding component of cell membrane invaginations (caveolae). It is involved in sequestering numerous effectors and signaling molecules and has antiapototic activities in prostate cancer. Perineural invasion (PNI) is associated with decreased apoptosis of cancer cells both in human tissues and the in vitro PNI model. We show here that stromal (perineurium) production of cav-1 is involved in a paracrine antiapoptotic loop in PNI. Transforming growth factor-beta1 is up-regulated in the cancer cells as they approach the nerve and is thought to up-regulate cav-1 in the perineurium of nerves with prostate cancer. Cav-1 is then secreted into the microenvironment and used by prostate cancer cells to inhibit apoptosis. In the in vitro PNI model, this phenomenon is partially reversed by neutralizing cav-1 antibodies or using ganglia from cav-1 knockout mice. Our results show a novel paracrine mechanism used by the prostate cancer in PNI to increase their proliferative activity and decrease apoptosis.


Subject(s)
Apoptosis/physiology , Caveolin 1/physiology , Prostate/innervation , Prostatic Neoplasms/pathology , Animals , Antibodies/pharmacology , Caveolin 1/biosynthesis , Caveolin 1/genetics , Caveolin 1/immunology , Cell Line, Tumor , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peripheral Nerves/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Stromal Cells/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , Up-Regulation
13.
Mol Cell Biol ; 23(24): 9389-404, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14645548

ABSTRACT

Previously it has been reported that caveolin-1 (cav-1) has antiapoptotic activities in prostate cancer cells and functions downstream of androgenic stimulation. In this study, we demonstrate that cav-1 overexpression significantly reduced thapsigargin (Tg)-stimulated apoptosis. Examination of the phosphatidylinositol 3-kinase (PI3-K)/Akt signaling cascade revealed higher activities of PDK1 and Akt but not PI3-K in cav-1-stimulated cells compared to control cells. We subsequently found that cav-1 interacts with and inhibits serine/threonine protein phosphatases PP1 and PP2A through scaffolding domain binding site interactions. Deletion of the cav-1 scaffolding domain significantly reduces phosphorylated Akt and cell viability compared with wild-type cav-1. Analysis of potential substrates for PP1 and PP2A revealed that cav-1-mediated inhibition of PP1 and PP2A leads to increased PDK1, Akt, and ERK1/2 activities. We demonstrate that increased Akt activities are largely responsible for cav-1-mediated cell survival using dominant-negative Akt mutants and specific inhibitors to MEK1/MEK and show that cav-1 increases the half-life of phosphorylated PDK1 and Akt after inhibition of PI3-K by LY294002. We further demonstrate that cav-1-stimulated Akt activities lead to increased phosphorylation of multiple Akt substrates, including GSK3, FKHR, and MDM2. In addition, overexpression of cav-1 significantly increases translocation of phosphorylated androgen receptor to nucleus. Our studies therefore reveal a novel mechanism of Akt activation in prostate cancer and potentially other malignancies.


Subject(s)
Caveolins/metabolism , Phosphoprotein Phosphatases/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Active Transport, Cell Nucleus , Binding Sites , Caveolin 1 , Caveolins/chemistry , Caveolins/genetics , Cell Death/drug effects , Cell Line, Tumor , Enzyme Activation , Humans , In Vitro Techniques , Male , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphorylation , Prostatic Neoplasms/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thapsigargin/pharmacology , Transfection
14.
Clin Cancer Res ; 9(10 Pt 1): 3653-9, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-14506154

ABSTRACT

PURPOSE: Caveolin-1 (cav-1), the major protein component of caveolae, plays an important role in multiple signaling pathways, molecular transport, and cellular proliferation and differentiation. The specific functions of cav-1/caveolae are highly cell and context dependent. We have previously shown that cav-1 expression is increased in metastatic human prostate cancer and that cav-1 cellular protein expression is predictive of recurrence of the disease after radical prostatectomy. Recently, we reported that cav-1 is secreted by androgen-insensitive prostate cancer cells, and we detected, by Western blotting, cav-1 in the high-density lipoprotein(3) fraction of serum specimens from patients with prostate cancer. EXPERIMENTAL DESIGN: Using rabbit polyclonal antibodies with specificity for cav-1, we developed a direct sandwich immunoassay for the determination of cav-1 in serum. A recombinant human cav-1 fusion protein was overexpressed and purified from 293 PE cells and used as a calibrator. RESULTS: The assay was highly specific and had a minimum detection limit of 0.017 ng/ml (mean + 3 SD of zero calibrator) and measuring range of up to 200 ng/ml. Intra-assay coefficient of variation was 2.29-6.74% and inter-assay coefficient of variation was 2.81-6.43% over the serum concentration tested 0.04-31.89 ng/ml. The recovery limit of cav-1 by the assay was 89.55-100.28%. The median serum cav-1 level in 102 prostate cancer patients with clinically localized disease (0.463 ng/ml) was significantly higher than 81 healthy control men (0.324 ng/ml; P = 0.0446, Mann-Whitney test) or 107 men with benign prostatic hyperplasia (0.172 ng/ml; P = 0.0317, Mann-Whitney test). CONCLUSIONS: Our results indicate that serum cav-1 has the power to differentiate between prostate cancer and benign prostatic hyperplasia patients and the potential to be an important biomarker for prostate cancer. Additional studies to test the potential of serum cav-1 as a diagnostic and/or prognostic marker in prostate cancer are warranted.


Subject(s)
Biomarkers, Tumor , Caveolins/blood , Enzyme-Linked Immunosorbent Assay/methods , Immunoassay/methods , Prostatic Neoplasms/blood , Aged , Animals , Antibodies/chemistry , Blotting, Western , Calibration , Caveolin 1 , Cell Line , Dose-Response Relationship, Drug , Humans , Male , Middle Aged , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/diagnosis , Rabbits , Recombinant Proteins/chemistry , Sensitivity and Specificity
15.
Curr Drug Targets ; 4(3): 251-61, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12643475

ABSTRACT

Prostate cancer is the most commonly diagnosed non-cutaneous cancer in adult males. Although prostate cancer that is confined to the gland can be cured in many patients using surgery or radiation, these treatments are only effective for localized tumors and the long-term failure rates for these treatments suggests that prostate cancer can metastasize relatively early in the course of the disease. Once prostate cancer has metastasized there are no curative therapies. The greatest challenge in the treatment of advanced prostate cancer is to access and eliminate metastatic cells. Therefore, effective prostate cancer therapy will require novel strategies to target cancer cells both at the site of the primary tumor and at distant metastatic sites. In this article we review several therapeutic targets and approaches that may provide new treatments for metastatic prostate cancer. We discuss the use of small molecules to target specific molecular events associated with metastatic prostate cancer, the use of specific antibodies that target unique metastasis associated molecules and the use of various gene therapy strategies to achieve anti-metastatic activities.


Subject(s)
Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Animals , Antibodies/chemistry , Antibodies/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/immunology , Cancer Vaccines/therapeutic use , Genetic Therapy/methods , Humans , Immunotherapy , Male , Neoplasm Metastasis , Prostatic Neoplasms/immunology
16.
J Urol ; 168(4 Pt 1): 1589-96, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12352463

ABSTRACT

PURPOSE: We summarize the literature regarding androgen insensitive prostate cancer and caveolin-1. Caveolin-1 is a major structural component of caveolae, membrane micro-domains known to have important roles in signal transduction and lipid transport. MATERIALS AND METHODS: A review of the literature relevant to androgen insensitive caveolin-1 and prostate cancer included the first published report in 1998 through those published in March 2002. RESULTS: Caveolin-1 expression is increased in primary and metastatic human prostate cancer with highest levels observed after androgen ablation therapy. Recent studies have documented that caveolin-1 is secreted by prostate cancer cells and can be detected in the serum of men with prostate cancer. CONCLUSIONS: The results presented in this review establish that caveolin-1 is an autocrine/paracrine factor associated with androgen insensitive prostate cancer. They show the potential for caveolin-1 as a biomarker therapeutic target for this important malignancy.


Subject(s)
Caveolins/physiology , Prostatic Neoplasms/pathology , Animals , Biomarkers, Tumor/physiology , Caveolin 1 , Humans , Male , Neoplasm Metastasis/physiopathology
17.
Mol Cell Biol ; 22(10): 3345-57, 2002 May.
Article in English | MEDLINE | ID: mdl-11971968

ABSTRACT

We identified a novel mouse gene, mRTVP-1, as a p53 target gene using differential display PCR and extensive promoter analysis. The mRTVP-1 protein has 255 amino acids and differs from the human RTVP-1 (hRTVP-1) protein by two short in-frame deletions of two and nine amino acids. RTVP-1 mRNA was induced in multiple cancer cell lines by adenovirus-mediated delivery of p53 and by gamma irradiation or doxorubicin both in the presence and in the absence of endogenous p53. Analysis of RTVP-1 expression in nontransformed and transformed cells further supported p53-independent gene regulation. Using luciferase reporter and electrophoretic mobility shift assays we identified a p53 binding site within intron 1 of the mRTVP-1 gene. Overexpression of mRTVP-1 or hRTVP-1 induced apoptosis in multiple cancer cell lines including prostate cancer cell lines 148-1PA, 178-2BMA, PC-3, TSU-Pr1, and LNCaP, a human lung cancer cell line, H1299, and two isogenic human colon cancer cell lines, HCT116 p53(+/+) and HCT116 p53(-/-), as demonstrated by annexin V positivity, phase-contrast microscopy, and in selected cases 4',6'-diamidino-2-phenylindole staining and DNA fragmentation. Deletion of the signal peptide from the N terminus of RTVP-1 reduced its apoptotic activities, suggesting that a secreted and soluble form of RTVP-1 may mediate, in part, its proapoptotic activities.


Subject(s)
Apoptosis/genetics , Neoplasm Proteins/metabolism , Promoter Regions, Genetic , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Animals , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Cell Size , Doxorubicin/pharmacology , Gamma Rays , Genes , Genes, Reporter , Humans , Membrane Proteins , Mice , Microscopy, Fluorescence , Molecular Sequence Data , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Polymerase Chain Reaction/methods , Protein Binding , Protein Sorting Signals , Sequence Alignment , Tumor Cells, Cultured , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...