Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Mol Cancer Res ; 20(1): 62-76, 2022 01.
Article in English | MEDLINE | ID: mdl-34610962

ABSTRACT

Although the Sonic hedgehog (SHH) signaling pathway has been implicated in promoting malignant phenotypes of prostate cancer, details on how it is activated and exerts its oncogenic role during prostate cancer development and progression is less clear. Here, we show that GLI3, a key SHH pathway effector, is transcriptionally upregulated during androgen deprivation and posttranslationally stabilized in prostate cancer cells by mutation of speckle-type POZ protein (SPOP). GLI3 is a substrate of SPOP-mediated proteasomal degradation in prostate cancer cells and prostate cancer driver mutations in SPOP abrogate GLI3 degradation. Functionally, GLI3 is necessary and sufficient for the growth and migration of androgen receptor (AR)-positive prostate cancer cells, particularly under androgen-depleted conditions. Importantly, we demonstrate that GLI3 physically interacts and functionally cooperates with AR to enrich an AR-dependent gene expression program leading to castration-resistant growth of xenografted prostate tumors. Finally, we identify an AR/GLI3 coregulated gene signature that is highly correlated with castration-resistant metastatic prostate cancer and predictive of disease recurrence. Together, these findings reveal that hyperactivated GLI3 promotes castration-resistant growth of prostate cancer and provide a rationale for therapeutic targeting of GLI3 in patients with castration-resistant prostate cancer (CRPC). IMPLICATIONS: We describe two clinically relevant mechanisms leading to hyperactivated GLI3 signaling and enhanced AR/GLI3 cross-talk, suggesting that GLI3-specific inhibitors might prove effective to block prostate cancer development or delay CRPC.


Subject(s)
Nerve Tissue Proteins/metabolism , Nuclear Proteins , Prostatic Neoplasms, Castration-Resistant/genetics , Repressor Proteins/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Zinc Finger Protein Gli3/metabolism , Animals , Cell Line, Tumor , Humans , Male , Mice , Mutation , Receptors, Androgen/metabolism
2.
Int J Mol Sci ; 22(4)2021 Feb 12.
Article in English | MEDLINE | ID: mdl-33673346

ABSTRACT

2-Methoxyestradiol (2-ME2) possesses anti-tumorigenic activities in multiple tumor models with acceptable tolerability profile in humans. Incomplete understanding of the mechanism has hindered its development as an anti-tumorigenic compound. We have identified for the first-time macrophage stimulatory protein 1 receptor (MST1R) as a potential target of 2-ME2 in prostate cancer cells. Human tissue validation studies show that MST1R (a.k.a RON) protein levels are significantly elevated in prostate cancer tissues compared to adjacent normal/benign glands. Serum levels of macrophage stimulatory protein (MSP), a ligand for RON, is not only associated with the risk of disease recurrence, but also significantly elevated in samples from African American patients. 2-ME2 treatment inhibited mechanical properties such as adhesion and elasticity that are associated with epithelial mesenchymal transition by downregulating mRNA expression and protein levels of MST1R in prostate cancer cell lines. Intervention with 2-ME2 significantly reduced tumor burden in mice. Notably, global metabolomic profiling studies identified significantly higher circulating levels of bile acids in castrated animals that were decreased with 2-ME2 intervention. In summary, findings presented in this manuscript identified MSP as a potential marker for predicting biochemical recurrence and suggest repurposing 2-ME2 to target RON signaling may be a potential therapeutic modality for prostate cancer.


Subject(s)
2-Methoxyestradiol/pharmacology , Drug Repositioning , Neoplasm Proteins , Prostatic Neoplasms , Receptor Protein-Tyrosine Kinases , Animals , Humans , Male , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , PC-3 Cells , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism
3.
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
4.
PLoS One ; 14(12): e0226187, 2019.
Article in English | MEDLINE | ID: mdl-31856170

ABSTRACT

The purpose of this investigation was to compare the antitumorigenic effects of the natural product Nexrutine to voluntary wheel running (VWR) in the transgenic adenocarcinoma of the mouse prostate (TRAMP) model. Forty-five, 10-week old TRAMP mice were randomized to either receive free access to the running wheel, Nexrutine pelleted into chow at 600 mg/kg or no treatment control. Mice were serially sacrificed at weeks 4, 8,12 and 20 weeks. Palpable tumors, body weight, food consumption and running wheel activity were monitored weekly. At necropsy, tumors and serum were harvested and stored for analysis. Serum was used to quantify circulating cytokines in 4 and 20 week time points. Nexrutine supplementation led to a 66% protection against high grade tumors. Exercise resulted in a 60% protection against high grade tumors. Both interventions reduced concentrations of IL-1α. Exercise also significantly lowered concentrations of eotaxin, IL-5, IL-12(p40) and VEGF. While there were no significant differences at baseline, exercise mice had significantly lower IL-5 and VEGF compared to control at the 20 week time point. Nexrutine also significantly reduced circulating IL-9 concentrations. No significant differences were observed when compared to the control group. Immunohistochemistry of tumor sections showed significantly lower expression of pAkt in Nexrutine fed mice with no visible differences for NFκB. In conclusion, both Nexrutine and exercise suppressed tumor growth. Though similar outcomes were seen in this comparative effectiveness study, the mechanisms by which exercise and Nexrutine exert this benefit may focus on different pathways.


Subject(s)
Physical Conditioning, Animal , Plant Extracts/pharmacology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/prevention & control , Animals , Cell Proliferation/drug effects , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Male , Mice , Mice, Transgenic , Neoplasm Grading , Phosphoproteins/metabolism , Prostatic Neoplasms/physiopathology , Proto-Oncogene Proteins c-akt/metabolism
5.
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
6.
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
7.
Int J Mol Sci ; 19(7)2018 Jul 20.
Article in English | MEDLINE | ID: mdl-30036938

ABSTRACT

COBRA1 (co-factor of BRCA1) is one of the four subunits of the negative elongation factor originally identified as a BRCA1-interacting protein. Here, we provide first-time evidence for the oncogenic role of COBRA1 in prostate pathogenesis. COBRA1 is aberrantly expressed in prostate tumors. It positively influences androgen receptor (AR) target gene expression and promoter activity. Depletion of COBRA1 leads to decreased cell viability, proliferation, and anchorage-independent growth in prostate cancer cell lines. Conversely, overexpression of COBRA1 significantly increases cell viability, proliferation, and anchorage-independent growth over the higher basal levels. Remarkably, AR-positive androgen dependent (LNCaP) cells overexpressing COBRA1 survive under androgen-deprivation conditions. Remarkably, treatment of prostate cancer cells with well-studied antitumorigenic agent, 2-methoxyestradiol (2-ME2), caused significant DNA methylation changes in 3255 genes including COBRA1. Furthermore, treatment of prostate cancer cells with 2-ME2 downregulates COBRA1 and inhibition of prostate tumors in TRAMP (transgenic adenocarcinomas of mouse prostate) animals with 2-ME2 was also associated with decreased COBRA1 levels. These observations implicate a novel role for COBRA1 in progression to CRPC and suggest that COBRA1 downregulation has therapeutic potential.


Subject(s)
BRCA1 Protein/metabolism , Nuclear Proteins/metabolism , Prostatic Neoplasms/metabolism , Transcription Factors/metabolism , 2-Methoxyestradiol , Animals , BRCA1 Protein/genetics , Cell Proliferation/genetics , Cell Proliferation/physiology , Cell Survival/genetics , Cell Survival/physiology , DNA Methylation/drug effects , DNA Methylation/genetics , Estradiol/analogs & derivatives , Estradiol/pharmacology , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Male , Mice , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Protein Binding , RNA-Binding Proteins , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Transcription Factors/genetics
8.
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
9.
Oncotarget ; 8(9): 15114-15125, 2017 Feb 28.
Article in English | MEDLINE | ID: mdl-28122346

ABSTRACT

Non-muscle invasive bladder cancer has a high recurrence rate of 45-70%, progressing to muscle invasive disease in about 15% of those patients over a 5-year period. Administration of the mycobacterium, Bacillus Calmette-Guerin (BCG) that induces local inflammation resulting in tumor remission in responsive patients is frequently used for treatment. BCG-treated patients with NF-κB del/del genotype have an increased risk of recurrence suggesting an important role of NF-κB in bladder cancer. Since protein methyltransferases play critical roles in modulating chromatin structure and gene expression, we screened a focused array of epigenetic modification genes to identify differential expression between normal urothelial and bladder cancer cells. We found and validated high expression of the SET-domain-containing protein methyltransferase, SETD6. SETD6 monomethylates NF-κB-p65 at lysine 310. Our results show that primary urothelial cells and normal bladder tissue have nearly undetectable message and protein level of SETD6 that increases in transformed urothelial cells and is further increased in bladder cancer cells and tissues. Overexpression of SETD6 in transformed urothelial cells increased cell survival and colony formation while knockdown in cancer cells decreased both parameters. Luciferase reporter assays showed that SETD6 induced the canonical NF-κB signaling pathway. Further, the use of catalytic SETD6 and IκBα mutant shows that SETD6 positively regulates survival by affecting p65 message, protein level and its function as determined by increased expression of NF-κB target genes. Our findings suggest that SETD6 plays an important role in NF-κB regulation and may have an important role in NF-κB-mediated local inflammatory response following BCG treatment.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , NF-kappa B/metabolism , Protein Methyltransferases/metabolism , Urinary Bladder Neoplasms/pathology , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Humans , NF-kappa B/genetics , Protein Methyltransferases/genetics , Signal Transduction , Tumor Cells, Cultured , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism
10.
Oncotarget ; 7(12): 14048-63, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26872377

ABSTRACT

Castrate-resistant prostate cancer (CRPC) is the fatal form of prostate cancer. Although reactivation of androgen receptor (AR) occurs following androgen deprivation, the precise mechanism involved is unclear. Here we show that the receptor tyrosine kinase, RON alters mechanical properties of cells to influence epithelial to mesenchymal transition and functions as a transcription factor to differentially regulate AR signaling. RON inhibits AR activation and subset of AR-regulated transcripts in androgen responsive LNCaP cells. However in C4-2B, a castrate-resistant sub-line of LNCaP and AR-negative androgen independent DU145 cells, RON activates subset of AR-regulated transcripts. Expression of AR in PC-3 cells leads to activation of RON under androgen deprivation but not under androgen proficient conditions implicating a role for RON in androgen independence. Consistently, RON expression is significantly elevated in castrate resistant prostate tumors. Taken together our results suggest that RON activation could aid in promoting androgen independence and that inhibition of RON in combination with AR antagonist(s) merits serious consideration as a therapeutic option during hormone deprivation therapy.


Subject(s)
Biomarkers, Tumor/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Androgen/metabolism , Apoptosis , Cell Proliferation , Humans , Male , Prognosis , Prostatic Neoplasms, Castration-Resistant/metabolism , Signal Transduction , Tumor Cells, Cultured
11.
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
12.
Clin Cancer Res ; 20(5): 1259-73, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24520096

ABSTRACT

PURPOSE: Near equal rates of incidence and mortality emphasize the need for novel targeted approaches for better management of patients with pancreatic cancer. Inflammatory molecules NF-κB and STAT3 are overexpressed in pancreatic tumors. Inhibition of one protein allows cancer cells to survive using the other. The goal of this study is to determine whether targeting STAT3/NF-κB crosstalk with a natural product Nexrutine can inhibit inflammatory signaling in pancreatic cancer. EXPERIMENTAL DESIGN: HPNE, HPNE-Ras, BxPC3, Capan-2, MIA PaCa-2, and AsPC-1 cells were tested for growth, apoptosis, cyclooxygenase-2 (COX-2), NF-κB, and STAT3 level in response to Nexrutine treatment. Transient expression, gel shift, chromatin immunoprecipitation assay was used to examine transcriptional regulation of COX-2. STAT3 knockdown was used to decipher STAT3/NF-κB crosstalk. Histopathologic and immunoblotting evaluation was performed on BK5-COX-2 transgenic mice treated with Nexrutine. In vivo expression of prostaglandin receptor E-prostanoid 4 (EP4) was analyzed in a retrospective cohort of pancreatic tumors using a tissue microarray. RESULTS: Nexrutine treatment inhibited growth of pancreatic cancer cells through induction of apoptosis. Reduced levels and activity of STAT3, NF-κB, and their crosstalk led to transcriptional suppression of COX-2 and subsequent decreased levels of prostaglandin E2 (PGE2) and PGF2. STAT3 knockdown studies suggest STAT3 as negative regulator of NF-κB activation. Nexrutine intervention reduced the levels of NF-κB, STAT3, and fibrosis in vivo. Expression of prostaglandin receptor EP4 that is known to play a role in fibrosis was significantly elevated in human pancreatic tumors. CONCLUSIONS: Dual inhibition of STAT3-NF-κB by Nexrutine may overcome problems associated with inhibition of either pathway.


Subject(s)
Cyclooxygenase 2/metabolism , NF-kappa B/metabolism , Pancreatic Neoplasms/metabolism , Plant Extracts/pharmacology , Receptors, Prostaglandin E, EP4 Subtype/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Fibrosis , Gene Expression , Humans , Immunohistochemistry , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Plant Extracts/administration & dosage , Protein Binding , Xenograft Model Antitumor Assays
13.
Cell Cycle ; 13(5): 845-58, 2014.
Article in English | MEDLINE | ID: mdl-24419204

ABSTRACT

Kaposi sarcoma-associated herpesvirus (KSHV) is a tumor virus encoding several proto-oncogenes. However, the roles of these viral genes in KSHV-induced tumorigenesis have not been defined. In this study, we used a recently developed model of KSHV-induced cellular transformation and tumorigenesis combining with a reverse genetic system to examine the role of a KSHV latent gene vCyclin (ORF72), a cellular Cyclin D2 homolog, in KSHV-induced oncogenesis. Deletion of vCyclin did not affect cell proliferation and cell cycle progression at a low-density condition, when cells were at an active proliferation state. However, vCyclin mutant cells were contact-inhibited and arrested at G 1 phase at a high-density condition. As a result, vCyclin mutant cells formed less and smaller colonies in soft agar assay. Nude mice inoculated with vCyclin mutant cells had reduced tumor incidence and extended tumor latency and survival compared with mice inoculated with wild-type (WT) virus-infected cells. WT but not mutant virus effectively induced Cyclin-dependent kinase inhibitor p27/Kip1 Ser10 phosphorylation and cytoplasmic relocalization. shRNA knockdown of p27 released the blockage of the mutant cells from cell cycle arrest at G 1 phase at a high-density condition. Together, these results indicate that vCyclin primarily functions to enhance cellular transformation and tumorigenesis by promoting cell cycle progression and cell proliferation at a contact-inhibited condition.


Subject(s)
Carcinogenesis/genetics , Contact Inhibition/physiology , Cyclin D2/metabolism , Herpesvirus 8, Human/physiology , Viral Proteins/metabolism , Animals , Apoptosis , Carcinogenesis/pathology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Cellular Senescence , Cyclin D2/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Mice, Nude , Mutation , Phosphorylation , Viral Proteins/genetics
14.
Cancer Prev Res (Phila) ; 6(1): 27-39, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23248098

ABSTRACT

SIRT1 (mammalian ortholog of the yeast silent information regulator 2) is a NAD-dependent histone deacetylase belonging to the multigene family of sirtuins. Anecdotal and epidemiologic observations provide evidence for beneficial effects of the calorie restriction mimetic resveratrol (RES), a SIRT1 activator in preventing cardiovascular diseases and cancer. Although SIRT1 possesses both tumorigenic and antitumorigenic potential, the molecular mechanisms underlying SIRT1-mediated tumor progression or inhibition are poorly understood. In this study, we investigated the role of SIRT1 in multiple human prostate cancer cell lines and prostate-specific PTEN knockout mouse model using resveratrol. Androgen-independent prostate cancer cell lines (C42B, PC3, and DU145) express higher levels of SIRT1 than androgen-responsive (LNCaP) and nontumorigenic prostate cells (RWPE-1). Resveratrol enhanced this expression without any significant effect on SIRT1 enzymatic activity. Inhibition of SIRT1 expression using shRNA enhanced cell proliferation and inhibited autophagy by repressing phosphorylation of S6K and 4E-BP1. These biologic correlates were reversed in the presence of resveratrol. Analysis of prostates from dietary intervention with resveratrol showed a significant reduction in prostate weight and reduction in the incidence of high-grade prostatic intraepithelial neoplastic (HGPIN) lesions by approximately 54% with no significant change in body weight. Consistent with the in vitro findings, resveratrol intervention in the PTEN knockout mouse model was associated with reduction in the prostatic levels of mTOR complex 1 (mTORC1) activity and increased expression of SIRT1. These data suggest that SIRT1/S6K-mediated inhibition of autophagy drives prostate tumorigenesis. Therefore, modulation of SIRT1/S6K signaling represents an effective strategy for prostate cancer prevention.


Subject(s)
Prostatic Intraepithelial Neoplasia/drug therapy , Prostatic Neoplasms/drug therapy , Ribosomal Protein S6 Kinases/metabolism , Sirtuin 1/metabolism , Stilbenes/administration & dosage , Animal Feed , Animals , Cell Line, Tumor , Cell Survival , Diet , Humans , Immunohistochemistry/methods , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes/metabolism , Mutation , Phosphorylation , Prostatic Intraepithelial Neoplasia/prevention & control , Prostatic Neoplasms/prevention & control , RNA, Small Interfering/metabolism , Resveratrol , Signal Transduction , Stilbenes/pharmacology , TOR Serine-Threonine Kinases/metabolism , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Time Factors
15.
PLoS Pathog ; 9(12): e1003857, 2013.
Article in English | MEDLINE | ID: mdl-24385912

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is causally linked to several human cancers, including Kaposi's sarcoma, primary effusion lymphoma and multicentric Castleman's disease, malignancies commonly found in HIV-infected patients. While KSHV encodes diverse functional products, its mechanism of oncogenesis remains unknown. In this study, we determined the roles KSHV microRNAs (miRs) in cellular transformation and tumorigenesis using a recently developed KSHV-induced cellular transformation system of primary rat mesenchymal precursor cells. A mutant with a cluster of 10 precursor miRs (pre-miRs) deleted failed to transform primary cells, and instead, caused cell cycle arrest and apoptosis. Remarkably, the oncogenicity of the mutant virus was fully restored by genetic complementation with the miR cluster or several individual pre-miRs, which rescued cell cycle progression and inhibited apoptosis in part by redundantly targeting IκBα and the NF-κB pathway. Genomic analysis identified common targets of KSHV miRs in diverse pathways with several cancer-related pathways preferentially targeted. These works define for the first time an essential viral determinant for KSHV-induced oncogenesis and identify NF-κB as a critical pathway targeted by the viral miRs. Our results illustrate a common theme of shared functions with hierarchical order among the KSHV miRs.


Subject(s)
Carcinogenesis/genetics , Cell Proliferation , Cell Transformation, Viral/genetics , Herpesvirus 8, Human/genetics , MicroRNAs/physiology , Animals , Cell Survival/genetics , Cells, Cultured , Gene Regulatory Networks , Gene Targeting , Humans , Microarray Analysis , Rats , Signal Transduction/genetics , Transcriptome
16.
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
17.
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
18.
J Clin Invest ; 122(3): 1076-81, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22293176

ABSTRACT

Infections by viruses are associated with approximately 12% of human cancer. Kaposi's sarcoma-associated herpesvirus (KSHV) is causally linked to several malignancies commonly found in AIDS patients. The mechanism of KSHV-induced oncogenesis remains elusive, due in part to the lack of an adequate experimental system for cellular transformation of primary cells. Here, we report efficient infection and cellular transformation of primary rat embryonic metanephric mesenchymal precursor cells (MM cells) by KSHV. Cellular transformation occurred at as early as day 4 after infection and in nearly all infected cells. Transformed cells expressed hallmark vascular endothelial, lymphatic endothelial, and mesenchymal markers and efficiently induced tumors in nude mice. KSHV established latent infection in MM cells, and lytic induction resulted in low levels of detectable infectious virions despite robust expression of lytic genes. Most KSHV-induced tumor cells were in a latent state, although a few showed heterogeneous expression of lytic genes. This efficient system for KSHV cellular transformation of primary cells might facilitate the study of growth deregulation mechanisms resulting from KSHV infections.


Subject(s)
Embryonic Stem Cells/cytology , Gene Expression Regulation , Herpesvirus 8, Human/metabolism , Mesenchymal Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Line , Cell Transformation, Viral , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Nude , Microcirculation , Open Reading Frames , Rats , Time Factors
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...