Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Commun Biol ; 3: 12, 2020.
Article in English | MEDLINE | ID: mdl-31909204

ABSTRACT

NAD[P]H:quinone oxidoreductase 1 (NQO1) regulates cell fate decisions in response to stress. Oxidative stress supports cancer maintenance and progression. Previously we showed that knockdown of NQO1 (NQO1low) prostate cancer cells upregulate pro-inflammatory cytokines and survival under hormone-deprived conditions. Here, we tested the ability of NQO1low cells to form tumors. We found NQO1low cells form aggressive tumors compared with NQO1high cells. Biopsy specimens and circulating tumor cells showed biochemical recurrent prostate cancer was associated with low NQO1. NQO1 silencing was sufficient to induce SMAD-mediated TGFß signaling and mesenchymal markers. TGFß treatment decreased NQO1 levels and induced molecular changes similar to NQO1 knockdown cells. Functionally, NQO1 depletion increased migration and sensitivity to oxidative stress. Collectively, this work reveals a possible new gatekeeper role for NQO1 in counteracting cellular plasticity in prostate cancer cells. Further, combining NQO1 with TGFß signaling molecules may serve as a better signature to predict biochemical recurrence.


Subject(s)
Cell Plasticity/genetics , NAD(P)H Dehydrogenase (Quinone)/genetics , Oxidative Stress , Prostatic Neoplasms/physiopathology , Transforming Growth Factor beta/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Nude , NAD(P)H Dehydrogenase (Quinone)/metabolism , Prostatic Neoplasms/genetics , Transforming Growth Factor beta/metabolism , Up-Regulation/physiology
2.
Mol Carcinog ; 58(9): 1701-1710, 2019 09.
Article in English | MEDLINE | ID: mdl-31124185

ABSTRACT

The general transcription factor E2F1 reportedly functions in a protumorigenic manner in several cancer models. We show that the genetic context of cancer cells influence E2F1's role to impede the protumorigenic role. Thirty to fifty percent of melanoma patients carry mutant BRAF with about 90% of mutant BRAF melanomas being V600E mutation. Tissue microarrays from melanoma patients were used to establish an association between E2F1 and BRAFV600E . We show for the first time that low E2F1 levels in BRAFV600E melanomas are associated with lymph node metastasis. Genetic manipulation of E2F1 in BRAFV600E and BRAFwt cells were used to determine its role in malignant melanoma progression by examining effects on migration and invasion. E2F1-mediated negative regulation of myosin light chain kinase (MYLK) increased migration and invasion in BRAFV600E cells by phosphorylating myosin light chain and increased stress fiber formation. We show that E2F1 inhibits extracellular signal-regulated kinase (ERK) activation in BRAFV600E cells and provide evidence for a negative feedback loop between E2F1 and ERK in these cells. This study shows for the first time that E2F1 has a cancer protective role in oncogenic BRAF-activated melanoma cells and that loss of E2F1 can allow disease progression through a novel mechanism of E2F1-mediated MYLK regulation. This study has implications for oncogenic BRAF-activated tumors and resistance to targeted oncogenic BRAF therapy.


Subject(s)
Cell Movement/genetics , E2F1 Transcription Factor/genetics , Melanoma/genetics , Melanoma/pathology , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Humans , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , MAP Kinase Signaling System/genetics , Mutation/genetics , Phosphorylation/genetics , Proto-Oncogene Proteins B-raf/genetics , Melanoma, Cutaneous Malignant
3.
Mol Carcinog ; 58(6): 854-861, 2019 06.
Article in English | MEDLINE | ID: mdl-30859654

ABSTRACT

Published evidence shows a correlation between several molecular markers and prostate cancer (PCa) progression including in African Americans (AAs) who are disproportionately affected. Our early detection efforts led to the identification of elevated levels of antiapoptotic protein, c-FLIP and its upstream regulatory factors such as androgen receptor (AR), recepteur d'origine nantais (RON), a receptor tyrosine kinase in human prostate tumors. The primary objective of this study was to explore whether these markers play a role in racial disparities using immunohistochemistry in prostatectomy samples from a cohort of AA, Hispanic Whites (HWs), and non-Hispanic Whites (NHWs). Bivariable and multivariable logistic regression analyses were used to identify a statistical association between molecular markers, possible correlation with risk factors including race, obesity, prostate-specific antigen (PSA) and disease aggressiveness. Further, changes in the levels and expression of these molecular markers were also evaluated using human PCa cell lines. We found significantly elevated levels of RON ( P = 0.0082), AR ( P = 0.0001), c-FLIP ( P = 0.0071) in AAs compared with HWs or NHWs. Furthermore, a higher proportion of HW and NHWs had a high Gleason score (>6) but not PSA as compared to AAs ( P = 0.032). In summary, our findings suggest that PSA was important in predicting aggressive disease for the cohort overall; however, high levels of RON may play a role in predisposing AA men to develop aggressive disease. Future research is needed using large datasets to confirm these findings and to explore whether all or any of these markers could aid in race-specific stratification of patients for treatment.


Subject(s)
Biomarkers, Tumor/metabolism , Prostatic Neoplasms/ethnology , Prostatic Neoplasms/surgery , Receptor Protein-Tyrosine Kinases/metabolism , Up-Regulation , Adult , Black or African American , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cell Line, Tumor , Cohort Studies , Gene Expression Regulation, Neoplastic , Hispanic or Latino , Humans , Logistic Models , Male , Middle Aged , Neoplasm Invasiveness , Prostatectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Androgen/metabolism , White People
4.
Cancer Lett ; 433: 232-241, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30003927

ABSTRACT

Radiation therapy (XRT) is a standard treatment for prostate cancer (PCa). Although dose escalation increases local control, toxicity hampers further escalation. Broader improvement will be possible by the addition of adjuvant therapies, which can synergize with radiation and thus improve efficacy. We have identified a natural compound (Nexrutine, Nx) that inhibits the survival and growth of PCa cells in combination with radiation. Combination studies demonstrated strong interaction between Nx and radiation both in vitro in multiple PCa cell lines and in the Transgenic adenocarcinoma of mouse prostate (TRAMP) model. Nx potentiated growth inhibitory effects of IR by down regulating ribosomal protein S6K (RPS6KB1), CyclinD1, Chk1 and HIF-1 α and prolonging G2/M checkpoint block. RPS6KB1 is upregulated in prostate cancers and its expression is correlated with tumor grade. Knockdown of RPS6KB1 in PCa cells increased their sensitivity toward radiation-induced survival inhibition. Overall, we provide scientific evidence (i) in support of Nx as an adjuvant in PCa patients receiving XRT (ii) suggesting that RPS6KB1 is an important player in Nx-mediated combinatorial benefits and emphasizes that RPS6KB1 is a novel target for PCa treatment. These data underscore the need to test the agent in additional preclinical models to validate these observations.


Subject(s)
Antineoplastic Agents/pharmacology , Plant Extracts/pharmacology , Prostatic Neoplasms/radiotherapy , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Checkpoint Kinase 1/biosynthesis , Cyclin D1/biosynthesis , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Male , Mice , PC-3 Cells , Ribosomal Protein S6 Kinases, 70-kDa/biosynthesis
5.
Cancer Res ; 74(19): 5644-55, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25125658

ABSTRACT

NADPH reductase NAD(P)H: quinone oxidoreductase 1 (NQO1) is needed to maintain a cellular pool of antioxidants, and this enzyme may contribute to tumorigenesis on the basis of studies in NQO1-deficient mice. In this work, we sought deeper insights into how NQO1 contributes to prostate carcinogenesis, a setting in which oxidative stress and inflammation are established contributors to disease development and progression. In the TRAMP mouse model of prostate cancer, NQO1 was highly expressed in tumor cells. NQO1 silencing in prostate cancer cells increased levels of nuclear IKKα and NF-κB while decreasing the levels of p53, leading to interactions between NF-κB and p300 that reinforce survival signaling. Gene expression analysis revealed upregulation of a set of immune-associated transcripts associated with inflammation and tumorigenesis in cells in which NQO1 was attenuated, with IL8 confirmed functionally in cell culture as one key NQO1-supported cytokine. Notably, NQO1-silenced prostate cancer cells were more resistant to androgen deprivation. Furthermore, NQO1 inhibition increased migration, including under conditions of androgen deprivation. These results reveal a molecular link between NQO1 expression and proinflammatory cytokine signaling in prostate cancer. Furthermore, our results suggest that altering redox homeostasis through NQO1 inhibition might promote androgen-independent cell survival via opposing effects on NF-κB and p53 function.


Subject(s)
Carcinogenesis , Inflammation Mediators/physiology , NAD(P)H Dehydrogenase (Quinone)/physiology , NF-kappa B/metabolism , Prostatic Neoplasms/physiopathology , p300-CBP Transcription Factors/metabolism , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Interleukin-8/biosynthesis , Male , NAD(P)H Dehydrogenase (Quinone)/genetics , Prostatic Neoplasms/pathology , Protein Binding , Real-Time Polymerase Chain Reaction , Tumor Suppressor Protein p53/metabolism
6.
PLoS One ; 7(9): e44917, 2012.
Article in English | MEDLINE | ID: mdl-23028678

ABSTRACT

Prediction of prostate cancer prognosis is challenging and predictive biomarkers of recurrence remain elusive. Although prostate specific antigen (PSA) has high sensitivity (90%) at a PSA level of 4.0 ng/mL, its low specificity leads to many false positive results and considerable overtreatment of patients and its performance at lower ranges is poor. Given the histopathological and molecular heterogeneity of prostate cancer, we propose that a panel of markers will be a better tool than a single marker. We tested a panel of markers composed of the anti-apoptotic protein FLIP and its transcriptional regulators Sp1 and Sp3 using prostate tissues from 64 patients with recurrent and non-recurrent cancer who underwent radical prostatectomy as primary treatment for prostate cancer and were followed with PSA measurements for at least 5 years. Immunohistochemical staining for Sp1, Sp3, and FLIP was performed on these tissues and scored based on the proportion and intensity of staining. The predictive value of the FLIP/Sp1/Sp3 signature for clinical outcome (recurrence vs. non-recurrence) was explored with logistic regression, and combinations of FLIP/Sp1/Sp3 and Gleason score were analyzed with a stepwise (backward and forward) logistic model. The discrimination of the markers was identified by sensitivity-specificity analysis and the diagnostic value of FLIP/Sp1/Sp3 was determined using area under the curve (AUC) for receiver operator characteristic curves. The AUCs for FLIP, Sp1, Sp3, and Gleason score for predicting PSA failure and non-failure were 0.71, 0.66, 0.68, and 0.76, respectively. However, this increased to 0.93 when combined. Thus, the "biomarker signature" of FLIP/Sp1/Sp3 combined with Gleason score predicted disease recurrence and stratified patients who are likely to benefit from more aggressive treatment.


Subject(s)
Biomarkers, Tumor/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/metabolism , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Aged , Analysis of Variance , Area Under Curve , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Prognosis , Prostatic Neoplasms/genetics , ROC Curve , Recurrence
7.
Endocr Relat Cancer ; 19(6): 759-77, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22993077

ABSTRACT

As prostate cancer (CaP) is regulated by androgen receptor (AR) activity, metastatic CaP is treated with androgen deprivation therapy (ADT). Despite initial response, patients on ADT eventually progress to castration-resistant CaP (CRPC), which is currently incurable. We previously showed that cleavage of the 280 kDa structural protein Filamin A (FlnA) to a 90 kDa fragment, and nuclear localization of the cleaved product, sensitized CRPC cells to ADT. Hence, treatment promoting FlnA nuclear localization would enhance androgen responsiveness. Here, we show that FlnA nuclear localization induced apoptosis in CRPC cells during ADT, identifying it as a treatment tool in advanced CaP. Significantly, the natural product genistein combined polysaccharide (GCP) had a similar effect. Investigation of the mechanism of GCP-induced apoptosis showed that GCP induced FlnA cleavage and nuclear localization and that apoptosis resulting from GCP treatment was mediated by FlnA nuclear localization. Two main components of GCP are genistein and daidzein: the ability of GCP to induce G2 arrest was due to genistein whereas sensitivity to ADT stemmed from daidzein; hence, both were needed to mediate GCP's effects. FlnA cleavage is regulated by its phosphorylation; we show that ADT enhanced FlnA phosphorylation, which prevented its cleavage, whereas GCP inhibited FlnA phosphorylation, thereby sensitizing CaP cells to ADT. In a mouse model of CaP recurrence, GCP, but not vehicle, impeded relapse following castration, indicating that GCP, when administered with ADT, interrupted the development of CRPC. These results demonstrate the efficacy of GCP in promoting FlnA nuclear localization and enhancing androgen responsiveness in CaP.


Subject(s)
Androgens/metabolism , Filamins/metabolism , Genistein/pharmacology , Polysaccharides/pharmacology , Prostatic Neoplasms/metabolism , Androgen Antagonists/pharmacology , Anilides/pharmacology , Animals , Apoptosis/drug effects , Castration , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Nucleus/metabolism , Genistein/therapeutic use , Male , Mice , Mice, Nude , Nitriles/pharmacology , Polysaccharides/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Tosyl Compounds/pharmacology , Tumor Burden
8.
PLoS Pathog ; 7(5): e1002054, 2011 May.
Article in English | MEDLINE | ID: mdl-21625536

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) establishes a latent infection in the host following an acute infection. Reactivation from latency contributes to the development of KSHV-induced malignancies, which include Kaposi's sarcoma (KS), the most common cancer in untreated AIDS patients, primary effusion lymphoma and multicentric Castleman's disease. However, the physiological cues that trigger KSHV reactivation remain unclear. Here, we show that the reactive oxygen species (ROS) hydrogen peroxide (H2O2) induces KSHV reactivation from latency through both autocrine and paracrine signaling. Furthermore, KSHV spontaneous lytic replication, and KSHV reactivation from latency induced by oxidative stress, hypoxia, and proinflammatory and proangiogenic cytokines are mediated by H2O2. Mechanistically, H2O2 induction of KSHV reactivation depends on the activation of mitogen-activated protein kinase ERK1/2, JNK, and p38 pathways. Significantly, H2O2 scavengers N-acetyl-L-cysteine (NAC), catalase and glutathione inhibit KSHV lytic replication in culture. In a mouse model of KSHV-induced lymphoma, NAC effectively inhibits KSHV lytic replication and significantly prolongs the lifespan of the mice. These results directly relate KSHV reactivation to oxidative stress and inflammation, which are physiological hallmarks of KS patients. The discovery of this novel mechanism of KSHV reactivation indicates that antioxidants and anti-inflammation drugs could be promising preventive and therapeutic agents for effectively targeting KSHV replication and KSHV-related malignancies.


Subject(s)
Herpesvirus 8, Human/physiology , Hydrogen Peroxide/metabolism , Reactive Oxygen Species/metabolism , Virus Activation , Virus Latency , Acetylcysteine/metabolism , Animals , Blotting, Western , Butadienes/pharmacology , Catalase/metabolism , Cell Line , Fluorescent Antibody Technique , Glutathione/metabolism , HEK293 Cells , Herpesviridae Infections , Herpesvirus 8, Human/metabolism , Humans , Imidazoles/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Nitriles/pharmacology , Oxidative Stress , Pyridines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Kaposi/virology , Signal Transduction , Virus Replication , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Cancer Res ; 70(14): 5994-6003, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20587519

ABSTRACT

Patients with advanced prostate cancer (PCa) are initially susceptible to androgen withdrawal (AW), but ultimately develop resistance to this therapy (castration-resistant PCa, CRPC). Here, we show that AW can promote CRPC development by increasing the levels of the receptor tyrosine kinase ErbB3 in androgen-dependent PCa, resulting in AW-resistant cell cycle progression and increased androgen receptor (AR) transcriptional activity. CRPC cell lines and human PCa tissue overexpressed ErbB3, whereas downregulation of ErbB3 prevented CRPC cell growth. Investigation of the mechanism by which AW augments ErbB3, using normal prostate-derived pRNS-1-1 cells, and androgen-dependent PCa lines LNCaP, PC346C, and CWR22 mouse xenografts, revealed that the AR suppresses ErbB3 protein levels, whereas AW relieves this suppression, showing for the first time the negative regulation of ErbB3 by AR. We show that AR activation promotes ErbB3 degradation in androgen-dependent cells, and that this effect is mediated by AR-dependent transcriptional upregulation of neuregulin receptor degradation protein-1 (Nrdp1), an E3 ubiquitin ligase that targets ErbB3 for degradation but whose role in PCa has not been previously examined. Therefore, AW decreases Nrdp1 expression, promoting ErbB3 protein accumulation, and leading to AR-independent proliferation. However, in CRPC sublines of LNCaP and CWR22, which strongly overexpress the AR, ErbB3 levels remain elevated due to constitutive suppression of Nrdp1, which prevents AR regulation of Nrdp1. Our observations point to a model of CRPC development in which progression of PCa to castration resistance is associated with the inability of AR to transcriptionally regulate Nrdp1, and predict that inhibition of ErbB3 during AW may impair CRPC development.


Subject(s)
Neoplasms, Hormone-Dependent/metabolism , Prostatic Neoplasms/metabolism , Receptor, ErbB-3/biosynthesis , Receptors, Androgen/metabolism , Ubiquitin-Protein Ligases/metabolism , Androgens/deficiency , Animals , Cell Growth Processes/physiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Nude , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Orchiectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism , Receptors, Androgen/genetics , Substance Withdrawal Syndrome/etiology , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/pathology , Transcriptional Activation , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
10.
Clin Cancer Res ; 15(3): 788-96, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19188148

ABSTRACT

PURPOSE: We previously showed that nuclear localization of the actin-binding protein, filamin A (FlnA), corresponded to hormone-dependence in prostate cancer. Intact FlnA (280 kDa, cytoplasmic) cleaved to a 90 kDa fragment which translocated to the nucleus in hormone-naïve cells, whereas in hormone-refractory cells, FlnA was phosphorylated, preventing its cleavage and nuclear translocation. We have examined whether FlnA localization determines a propensity to metastasis in advanced androgen-independent prostate cancer. EXPERIMENTAL DESIGN: We examined, by immunohistochemistry, FlnA localization in paraffin-embedded human prostate tissue representing different stages of progression. Results were correlated with in vitro studies in a cell model of prostate cancer. RESULTS: Nuclear FlnA was significantly higher in benign prostate (0.6612 +/- 0.5888), prostatic intraepithelial neoplasia (PIN; 0.6024 +/- 0.4620), and clinically localized cancers (0.69134 +/- 0.5686) compared with metastatic prostate cancers (0.3719 +/- 0.4992, P = 0.0007). Cytoplasmic FlnA increased from benign prostate (0.0833 +/- 0.2677), PIN (0.1409 +/- 0.2293), localized cancers (0.3008 +/- 0.3762, P = 0.0150), to metastases (0.7632 +/- 0.4414, P < 0.00001). Logistic regression of metastatic versus nonmetastatic tissue yielded the area under the receiver operating curve as 0.67 for nuclear-FlnA, 0.79 for cytoplasmic-FlnA, and 0.82 for both, indicating that metastasis correlates with cytoplasmic to nuclear translocation. In vitro studies showed that cytoplasmic localization of FlnA induced cell invasion whereas nuclear translocation of the protein inhibited it. FlnA dephosphorylation with the protein kinase A inhibitor H-89 facilitated FlnA nuclear translocation, resulting in decreased invasiveness and AR transcriptional activity, and induced sensitivity to androgen withdrawal in hormone-refractory cells. CONCLUSIONS: The data presented in this study indicate that in prostate cancer, metastasis correlates with cytoplasmic localization of FlnA and may be prevented by cleavage and subsequent nuclear translocation of this protein.


Subject(s)
Cell Nucleus/metabolism , Contractile Proteins/metabolism , Cytoplasm/metabolism , Microfilament Proteins/metabolism , Prostatic Neoplasms/metabolism , Androgen Antagonists/therapeutic use , Cell Line, Tumor , Cell Movement , Drug Resistance, Neoplasm , Filamins , Humans , Immunohistochemistry , Male , Neoplasm Invasiveness , Neoplasm Metastasis , Prostatic Neoplasms/pathology , Tissue Array Analysis
11.
Endocr Relat Cancer ; 12(1): 119-34, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15788644

ABSTRACT

In a previous report, we showed that increased activation of Akt, a downstream effector of phosphoinositide 3-kinase (PI3K) together with decreased activation of extracellular-signal-regulated kinase (ERK), a member of the mitogen-activated protein kinase (MAPK) family, predicted poor clinical outcome in prostate cancer (Kreisberg et al. 2004 Cancer Research 64 5232-5236). We now show that Akt activation, but not ERK activation, is correlated with proliferation in human prostate tumors as estimated by the expression of the cell proliferation antigen Ki67. We verified these results in vitro, using the androgen-dependent prostate cancer cell line LNCaP and its androgen-independent clone C4-2 as models of prostate cancer of good and poor clinical outcome, respectively. C4-2 cells expressed higher Akt activation, lower ERK activation and increased proliferation compared with LNCaP cells, similar to cases of poor clinical outcome. The PI3K inhibitor LY294002, but not the MAPK/ERK kinase inhibitor PD98059, induced growth arrest in both cell lines. Transient transfection with constitutively active Akt increased proliferation while dominant negative Akt decreased it, thus showing that Akt plays an important role in prostate cancer proliferation. Akt regulates the expression and activation of the androgen receptor. Androgen receptor inhibition with Casodex induced growth arrest in LNCaP cells, but not in C4-2 cells. Another PI3K downstream effector, p70 S6 kinase, requires prior phosphorylation by mammalian target of rapamycin (mTOR) for complete activation. Activation of p70 S6 kinase was higher in C4-2 compared with LNCaP cells. Rapamycin, an mTOR inhibitor, had a growth-inhibitory effect in C4-2 cells, but not in LNCaP cells. Our data suggest a shift from a Casodex-sensitive proliferation pathway in LNCaP cells to a rapamycin-sensitive pathway in C4-2 cells.


Subject(s)
Androgens/pharmacology , Cell Proliferation/drug effects , Neoplasms, Hormone-Dependent/metabolism , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Androgen Antagonists/pharmacology , Androgen Receptor Antagonists , Anilides/pharmacology , Antibiotics, Antineoplastic/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Ki-67 Antigen/metabolism , Male , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/secondary , Nitriles , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/secondary , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-akt , Receptors, Androgen/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Tosyl Compounds , Tumor Cells, Cultured
12.
Cancer Res ; 64(15): 5232-6, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15289328

ABSTRACT

We previously showed, by immunohistochemistry with phospho-specific antibodies, increased phosphorylation (activation) of Akt (Ser(473)) [phosphorylated Akt (pAkt)] in high-Gleason grade prostate cancer (Malik SN, et al., Clin Cancer Res 2002;8:1168-71). Elevation of pAkt was accompanied by decreased phosphorylation of extracellular signal-regulated kinase (ERK) 1/2 (Thr(202)/Tyr(204)) [phosphorylated ERK (pERK)], indicative of inactivation. In this report, we determined whether increased pAkt and decreased pERK predicted clinical outcome. Prostate-specific antigen (PSA) failure (detectable and rising PSA) versus PSA non-failure (undetectable PSA 5 years after prostatectomy) was used as a surrogate for clinical outcome. Prostate tumors from cases of PSA failure versus non-failure were stained for pAkt and pERK. A significant increase in mean pAkt staining (P < 0.001) in the PSA failures versus non-failures was seen based on the Wilcoxon signed ranks test [222.18 +/- 33.9 (n = 37) versus 108.79 +/- 104.57 (n = 16)]. Using the best-fitting multiple logistic regression equation, a 100-point increase in pAkt staining resulted in a 160% increase in the odds of being a PSA failure. There was decreased staining for pERK in PSA failures versus non-failures: a 100-point decrease resulted in an 80% increase in the odds of being a PSA failure. Each of these effects assumed the other biomarker was held constant. The area under the receiver-operating characteristic curve for these two biomarkers predicting PSA failure was 0.84, indicating excellent discrimination between PSA failure and non-failure cases. These data indicate that increased pAkt, alone or together with decreased pERK, is an important predictor of probability of PSA failure. However, pERK alone was not a significant predictor of PSA failure.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Humans , Immunoenzyme Techniques , Male , Middle Aged , Mitogen-Activated Protein Kinase 3 , Phosphorylation , Predictive Value of Tests , Prostate-Specific Antigen/metabolism , Prostatectomy , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt
SELECTION OF CITATIONS
SEARCH DETAIL
...