251 results on '"Tindall DJ"'
Search Results
2. Whole blood defensin mRNA expression is a predictive biomarker of docetaxel response in castration-resistant prostate cancer
- Author
-
Kohli M, Young CYF, Tindall DJ, Nandy D, McKenzie KM, Bevan GH, and Donkena KV
- Subjects
lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 - Abstract
Manish Kohli,1 Charles YF Young,2 Donald J Tindall,2 Debashis Nandy,1 Kyle M McKenzie,3 Graham H Bevan,4 Krishna Vanaja Donkena5 1Department of Oncology, 2Department of Urology, 3Department of Geriatric Medicine, Mayo Clinic, Rochester, MN, 4University of Rochester Medical Center, Rochester, NY, 5Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA Abstract: This study tested the potential of circulating RNA-based signals as predictive biomarkers for docetaxel response in patients with metastatic castration-resistant prostate cancer (CRPC). RNA was analyzed in blood from six CRPC patients by whole-transcriptome sequencing (total RNA-sequencing) before and after docetaxel treatment using the Illumina’s HiSeq platform. Targeted RNA capture and sequencing was performed in an independent cohort of ten patients with CRPC matching the discovery cohort to confirm differential expression of the genes. Response to docetaxel was defined on the basis of prostate-specific antigen levels and imaging criteria. Two-way analysis of variance was used to compare differential gene expression in patients classified as responders versus nonresponders before and after docetaxel treatment. Thirty-four genes with two-fold differentially expressed transcripts in responders versus nonresponders were selected from total RNA-sequencing for further validation. Targeted RNA capture and sequencing showed that 13/34 genes were differentially expressed in responders. Alpha defensin genes DEFA1, DEFA1B, and DEFA3 exhibited significantly higher expression in responder patients compared with nonresponder patients before administration of chemotherapy (fold change >2.5). In addition, post-docetaxel treatment significantly increased transcript levels of these defensin genes in responders (fold change >2.8). Our results reveal that patients with higher defensin RNA transcripts in blood respond well to docetaxel therapy. We suggest that monitoring DEFA1, DEFA1B, and DEFA3 RNA transcripts in blood prior to treatment will be helpful to determine which patients are better candidates to receive docetaxel chemotherapy. Keywords: docetaxel, RNA-seq, defensin, targeted RNA-seq, prostate cancer
- Published
- 2015
3. Modeling and analysis of Gleason score 8-10 prostate cancers in the REDUCE study
- Author
-
Andriole GL, Bostwick DG, Gomella LG, Marberger M, Tammela TL, Tindall DJ, Fowler IL, Garges HP, Wilson TH, Castro R., MONTORSI , FRANCESCO, Andriole, Gl, Bostwick, Dg, Gomella, Lg, Marberger, M, Montorsi, Francesco, Tammela, Tl, Tindall, Dj, Fowler, Il, Garges, Hp, Wilson, Th, and Castro, R.
- Published
- 2014
4. Characterization of a TPA-response element in hte 5'-flanking region of the androgen receptor gene
- Author
-
Kumar, MV, Jones, EA, Felts, SJ, Blexrud, MD, Grossmann, ME, Blok, Leen, Schmidt, LJ, Tindall, DJ, and Developmental Biology
- Published
- 1998
5. Mechanism of lymph node metastasis in prostate cancer.
- Author
-
Datta K, Muders M, Zhang H, Tindall DJ, Datta, Kaustubh, Muders, Michael, Zhang, Heyu, and Tindall, Donald J
- Abstract
Detection of lymph node metastases indicates poor prognosis for prostate cancer patients. Therefore, elucidation of the mechanism(s) of lymph node metastasis is important to understand the progression of prostate cancer and also to develop therapeutic interventions. In this article, the known mechanisms for lymph node metastasis are discussed and the involvement of lymphatic vessels in prostate cancer lymph node metastasis is comprehensively summarized. In addition, contradictory findings regarding the importance of lymphangiogenesis in facilitating lymph node metastasis in prostate cancer are pointed out and reconcilation is attempted. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
6. DHEA in elderly women and DHEA or testosterone in elderly men.
- Author
-
Nair KS, Rizza RA, O'Brien P, Dhatariya K, Short KR, Nehra A, Vittone JL, Klee GG, Basu A, Basu R, Cobelli C, Toffolo G, Dalla Man C, Tindall DJ, Melton LJ III, Smith GE, Khosla S, and Jensen MD
- Published
- 2006
7. Selective and Scalable Synthesis of Sugar Alcohols by Homogeneous Asymmetric Hydrogenation of Unprotected Ketoses.
- Author
-
Tindall DJ, Mader S, Kindler A, Rominger F, Hashmi ASK, and Schaub T
- Abstract
Sugar alcohols are of great importance for the food industry and are promising building blocks for bio-based polymers. Industrially, they are produced by heterogeneous hydrogenation of sugars with H
2 , usually with none to low stereoselectivities. Now, we present a homogeneous system based on commercially available components, which not only increases the overall yield, but also allows a wide range of unprotected ketoses to be diastereoselectively hydrogenated. Furthermore, the system is reliable on a multi-gram scale allowing sugar alcohols to be isolated in large quantities at high atom economy., (© 2020 Wiley-VCH GmbH.)- Published
- 2021
- Full Text
- View/download PDF
8. Ru 0 or Ru II : A Study on Stabilizing the "Activated" Form of Ru-PNP Complexes with Additional Phosphine Ligands in Alcohol Dehydrogenation and Ester Hydrogenation.
- Author
-
Tindall DJ, Menche M, Schelwies M, Paciello RA, Schäfer A, Comba P, Rominger F, Hashmi ASK, and Schaub T
- Abstract
The complex Ru-MACHO has been previously shown to undergo uncontrolled degradation subsequent to base-induced dehydrochlorination in the absence of a substrate. In this study, we report that stabilization of the dehydrochlorinated Ru-MACHO with phosphines furnishes complexes whose structures depend on the phosphines employed: while PMe
3 led to the expected octahedral RuII complex, PPh3 provided access to a trigonal-bipyramidal Ru0 complex. Because both complexes proved to be active in base-free (de)hydrogenation reactions, thorough quantum-chemical calculations were employed to understand the reaction mechanism. The calculations show that both complexes lead to the same mechanistic scenario after phosphine dissociation and, therefore, only differ energetically in this step. According to the calculations, the typically proposed metal-ligand cooperation mechanism is not the most viable pathway. Instead, a metal-ligand-assisted pathway is preferred. Finally, experiments show that phosphine addition enhances the catalyst's performance in comparison to the PR3 -free "activated" Ru-MACHO.- Published
- 2020
- Full Text
- View/download PDF
9. Exploring unsymmetrical diboranes(4) as boryl ligand precursors: platinum(ii) bis-boryl complexes.
- Author
-
Drescher Née Oschmann W, Borner C, Tindall DJ, and Kleeberg C
- Abstract
A series of five unsymmetrical platinum(ii) bis-boryl complexes, bearing two distinct boryl ligands, are obtained by the oxidative addition reaction of unsymmetrical diborane(4) derivatives, bearing either two different dialkoxy or one dialkoxy and one diamino boryl moiety, with [(Ph
3 P)2 Pt(C2 H4 )]. All five complexes were structurally and spectroscopically characterised. The bis-boryl platinum(ii) complexes exhibit slightly distorted square-planar cis -boryl structures with acute B-Pt-B angles, short B⋯B distances of 2.44-2.55 Å and relatively long trans -boryl P-Pt distances around 2.34 Å. The31 P-195 Pt NMR coupling constants are indicative for the strongly donating/ trans -influencing boryl ligands. Despite the structural and spectroscopic data at hand no finally conclusive order of the donor properties/ trans -influence of the boryl ligands can be deduced on the basis of these data. This may be explained by an (residual) interaction of two boryl ligands., Competing Interests: There are no conflicts to declare., (This journal is © The Royal Society of Chemistry.)- Published
- 2019
- Full Text
- View/download PDF
10. Androgen receptor splice variants bind to constitutively open chromatin and promote abiraterone-resistant growth of prostate cancer.
- Author
-
He Y, Lu J, Ye Z, Hao S, Wang L, Kohli M, Tindall DJ, Li B, Zhu R, Wang L, and Huang H
- Subjects
- Animals, Binding Sites, Cell Line, Tumor, Chromatin metabolism, Disease Progression, Drug Resistance, Neoplasm, Epigenesis, Genetic, Genomics, Humans, Male, Mice, SCID, Prostatic Neoplasms, Castration-Resistant drug therapy, Prostatic Neoplasms, Castration-Resistant metabolism, Prostatic Neoplasms, Castration-Resistant pathology, Protein Isoforms metabolism, RNA Splicing, Receptors, Androgen genetics, rap GTP-Binding Proteins physiology, Androstenes therapeutic use, Antineoplastic Agents therapeutic use, Prostatic Neoplasms, Castration-Resistant genetics, Receptors, Androgen metabolism
- Abstract
Androgen receptor (AR) splice variants (ARVs) are implicated in development of castration-resistant prostate cancer (CRPC). Upregulation of ARVs often correlates with persistent AR activity after androgen deprivation therapy (ADT). However, the genomic and epigenomic characteristics of ARV-dependent cistrome and the disease relevance of ARV-mediated transcriptome remain elusive. Through integrated chromatin immunoprecipitation coupled sequencing (ChIP-seq) and RNA sequencing (RNA-seq) analysis, we identified ARV-preferential-binding sites (ARV-PBS) and a set of genes preferentially transactivated by ARVs in CRPC cells. ARVs preferentially bind to enhancers located in nucleosome-depleted regions harboring the full AR-response element (AREfull), while full-length AR (ARFL)-PBS are enhancers resided in closed chromatin regions containing the composite FOXA1-nnnn-AREhalf motif. ARV-PBS exclusively overlapped with AR binding sites in castration-resistant (CR) tumors in patients and ARV-preferentially activated genes were up-regulated in abiraterone-resistant patient specimens. Expression of ARV-PBS target genes, such as oncogene RAP2A and cell cycle gene E2F7, were significantly associated with castration resistance, poor survival and tumor progression. We uncover distinct genomic and epigenomic features of ARV-PBS, highlighting that ARVs are useful tools to depict AR-regulated oncogenic genome and epigenome landscapes in prostate cancer. Our data also suggest that the ARV-preferentially activated transcriptional program could be targeted for effective treatment of CRPC.
- Published
- 2018
- Full Text
- View/download PDF
11. Structure and Reactivity of Half-Sandwich Rh(+3) and Ir(+3) Carbene Complexes. Catalytic Metathesis of Azobenzene Derivatives.
- Author
-
Tindall DJ, Werlé C, Goddard R, Philipps P, Farès C, and Fürstner A
- Abstract
Traditional rhodium carbene chemistry relies on the controlled decomposition of diazo derivatives with [Rh
2 (OAc)4 ] or related dinuclear Rh(+2) complexes, whereas the use of other rhodium sources is much less developed. It is now shown that half-sandwich carbene species derived from [Cp*MX2 ]2 (M = Rh, Ir; X = Cl, Br, I, Cp* = pentamethylcyclopentadienyl) also exhibit favorable application profiles. Interestingly, the anionic ligand X proved to be a critical determinant of reactivity in the case of cyclopropanation, epoxide formation and the previously unknown catalytic metathesis of azobenzene derivatives, whereas the nature of X does not play any significant role in -OH insertion reactions. This perplexing disparity can be explained on the basis of spectral and crystallographic data of a representative set of carbene complexes of this type, which could be isolated despite their pronounced electrophilicity. Specifically, the donor/acceptor carbene 10a derived from ArC(═N2 )COOMe and [Cp*RhCl2 ]2 undergoes spontaneous 1,2-migratory insertion of the emerging carbene unit into the Rh-Cl bond with formation of the C-metalated rhodium enolate 11. In contrast, the analogous complexes 10b,c derived from [Cp*RhX2 ]2 (X = Br, I) as well as the iridium species 13 and 14 derived from [Cp*IrCl2 ]2 are sufficiently stable and allow true carbene reactivity to be harnessed. These complexes are competent intermediates for the catalytic metathesis of azobenzene derivatives, which provides access to α-imino esters that would be difficult to make otherwise. Rather than involving metal nitrenes, the reaction proceeds via aza-ylides that evolve into diaziridines; a metastable compound of this type has been fully characterized.- Published
- 2018
- Full Text
- View/download PDF
12. Activation of P-TEFb by Androgen Receptor-Regulated Enhancer RNAs in Castration-Resistant Prostate Cancer.
- Author
-
Zhao Y, Wang L, Ren S, Wang L, Blackburn PR, McNulty MS, Gao X, Qiao M, Vessella RL, Kohli M, Zhang J, Karnes RJ, Tindall DJ, Kim Y, MacLeod R, Ekker SC, Kang T, Sun Y, and Huang H
- Subjects
- Base Sequence, Cell Line, Tumor, Cell Proliferation, Cyclin T metabolism, Enhancer Elements, Genetic genetics, Gene Expression Regulation, Neoplastic, Genetic Loci, Humans, Male, Models, Biological, Nucleotide Motifs genetics, Phosphorylation, Prostate-Specific Antigen genetics, Prostatic Neoplasms, Castration-Resistant pathology, Protein Binding, RNA Polymerase II metabolism, Serine metabolism, Up-Regulation genetics, Positive Transcriptional Elongation Factor B metabolism, Prostatic Neoplasms, Castration-Resistant genetics, RNA metabolism, Receptors, Androgen metabolism
- Abstract
The androgen receptor (AR) is required for castration-resistant prostate cancer (CRPC) progression, but the function and disease relevance of AR-bound enhancers remain unclear. Here, we identify a group of AR-regulated enhancer RNAs (e.g., PSA eRNA) that are upregulated in CRPC cells, patient-derived xenografts (PDXs), and patient tissues. PSA eRNA binds to CYCLIN T1, activates P-TEFb, and promotes cis and trans target gene transcription by increasing serine-2 phosphorylation of RNA polymerase II (Pol II-Ser2p). We define an HIV-1 TAR RNA-like (TAR-L) motif in PSA eRNA that is required for CYCLIN T1 binding. Using TALEN-mediated gene editing we further demonstrate that this motif is essential for increased Pol II-Ser2p occupancy levels and CRPC cell growth. We have uncovered a P-TEFb activation mechanism and reveal altered eRNA expression that is related to abnormal AR function and may potentially be a therapeutic target in CRPC., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
13. Androgen Receptor Upregulation Mediates Radioresistance after Ionizing Radiation.
- Author
-
Spratt DE, Evans MJ, Davis BJ, Doran MG, Lee MX, Shah N, Wongvipat J, Carnazza KE, Klee GG, Polkinghorn W, Tindall DJ, Lewis JS, and Sawyers CL
- Subjects
- Animals, Blotting, Western, Cell Line, Tumor, Comet Assay, Fluorescent Antibody Technique, Humans, Luminescent Measurements, Male, Mice, Mice, SCID, Prostatic Neoplasms metabolism, Real-Time Polymerase Chain Reaction, Up-Regulation, Xenograft Model Antitumor Assays, Prostatic Neoplasms radiotherapy, Radiation Tolerance physiology, Receptors, Androgen biosynthesis
- Abstract
Clinical trials have established the benefit of androgen deprivation therapy (ADT) combined with radiotherapy in prostate cancer. ADT sensitizes prostate cancer to radiotherapy-induced death at least in part through inhibition of DNA repair machinery, but for unknown reasons, adjuvant ADT provides further survival benefits. Here, we show that androgen receptor (AR) expression and activity are durably upregulated following radiotherapy in multiple human prostate cancer models in vitro and in vivo. Moreover, the degree of AR upregulation correlates with survival in vitro and time to tumor progression in animal models. We also provide evidence of AR pathway upregulation, measured by a rise in serum levels of AR-regulated hK2 protein, in nearly 20% of patients after radiotherapy. Furthermore, these men were three-fold more likely to experience subsequent biochemical failure. Collectively, these data demonstrate that radiotherapy can upregulate AR signaling after therapy to an extent that negatively affects disease progression and/or survival., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
14. Are androgen receptor variants a substitute for the full-length receptor?
- Author
-
Lu J, Van der Steen T, and Tindall DJ
- Subjects
- Alternative Splicing, Animals, Disease Progression, Gene Expression Profiling, Gene Expression Regulation physiology, Humans, Male, Neoplasms, Hormone-Dependent, Prostatic Neoplasms drug therapy, Prostatic Neoplasms genetics, Prostatic Neoplasms, Castration-Resistant metabolism, Protein Isoforms, Receptors, Androgen genetics, Signal Transduction physiology, Prostatic Neoplasms metabolism, Receptors, Androgen metabolism
- Abstract
Androgen receptor splice variants (AR-Vs)--which are expressed in castration-resistant prostate cancer (CRPC) cell lines and clinical samples--lack the C-terminal ligand-binding domain and are constitutively active. AR-Vs are, therefore, resistant to traditional androgen deprivation therapy (ADT). AR-Vs are induced by several mechanisms, including ADT, and might contribute to the progression of CRPC and resistance to ADT. AR-Vs could represent a novel therapeutic target for prostate cancer, especially in CRPC.
- Published
- 2015
- Full Text
- View/download PDF
15. The cistrome and gene signature of androgen receptor splice variants in castration resistant prostate cancer cells.
- Author
-
Lu J, Lonergan PE, Nacusi LP, Wang L, Schmidt LJ, Sun Z, Van der Steen T, Boorjian SA, Kosari F, Vasmatzis G, Klee GG, Balk SP, Huang H, Wang C, and Tindall DJ
- Subjects
- High-Throughput Nucleotide Sequencing, Humans, Male, Protein Isoforms, Tumor Cells, Cultured, Gene Expression Regulation, Neoplastic, Prostatic Neoplasms, Castration-Resistant genetics, Receptors, Androgen genetics, Transcriptome
- Abstract
Purpose: Spliced variant forms of androgen receptor were recently identified in castration resistant prostate cancer cell lines and clinical samples. We identified the cistrome and gene signature of androgen receptor splice variants in castration resistant prostate cancer cell lines and determined the clinical significance of androgen receptor splice variant regulated genes., Materials and Methods: The castration resistant prostate cancer cell line 22Rv1, which expresses full-length androgen receptor and androgen receptor splice variants endogenously, was used as the research model. We established 22Rv1-ARFL(-)/ARV(+) and 22Rv1-ARFL(-)/ARV(-) through RNA interference. Chromatin immunoprecipitation coupled with next generation sequencing and microarray techniques were used to identify the cistrome and gene expression profiles of androgen receptor splice variants in the absence of androgen., Results: Androgen receptor splice variant binding sites were identified in 22Rv1-ARFL(-)/ARV(+). A gene set was regulated uniquely by androgen receptor splice variants but not by full-length androgen receptor in the absence of androgen. Integrated analysis revealed that some genes were directly modulated by androgen receptor splice variants. Unsupervised clustering analysis showed that the androgen receptor splice variant gene signature differentiated benign from malignant prostate tissue as well as localized prostate cancer from metastatic castration resistant prostate cancer specimens. Some genes that were modulated uniquely by androgen receptor splice variants also correlated with histological grade and biochemical failure., Conclusions: Androgen receptor splice variants can bind to DNA independent of full-length androgen receptor in the absence of androgen and modulate a unique set of genes that is not regulated by full-length androgen receptor. The androgen receptor splice variant gene signature correlates with disease progression. It distinguishes primary cancer from castration resistant prostate cancer specimens and benign from malignant prostate specimens., (Copyright © 2015 American Urological Association Education and Research, Inc. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
16. Polycomb protein EZH2 suppresses apoptosis by silencing the proapoptotic miR-31.
- Author
-
Zhang Q, Padi SK, Tindall DJ, and Guo B
- Subjects
- Cell Line, Tumor, Docetaxel, Down-Regulation drug effects, Down-Regulation genetics, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Enhancer of Zeste Homolog 2 Protein, Gene Expression Regulation, Neoplastic drug effects, Gene Knockdown Techniques, Histones metabolism, Humans, Male, Methylation drug effects, MicroRNAs metabolism, Models, Biological, Promoter Regions, Genetic genetics, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, RNA, Small Interfering metabolism, Taxoids pharmacology, Apoptosis drug effects, Apoptosis genetics, Gene Silencing drug effects, MicroRNAs genetics, Polycomb Repressive Complex 2 metabolism
- Abstract
Enhancer of zeste homolog 2 (EZH2) is the catalytic subunit of the polycomb repressive complex 2 and suppresses gene expression by catalyzing histone H3 methylation on lysine 27. EZH2 is overexpressed in metastatic prostate cancer and has been shown to promote cell proliferation and metastasis. Here we show that EZH2 also suppresses prostate cancer apoptosis by coordinating the epigenetic silencing of two proapoptotic microRNAs (miRNA), miR-205 and miR-31. We previously reported that miR-205 promotes apoptosis by targeting antiapoptotic protein Bcl-w and miR-205 is silenced in prostate cancer through promoter methylation. In this study, we found that EZH2 suppresses miR-31 expression by trimethylation of lysine 27 on histone 3 on the miR-31 promoter. SiRNA knockdown of EZH2 increased miR-31 expression and decreased the antiapoptotic protein E2F6 (E2F transcription factor 6) (a target of miR-31), resulting in the sensitization of prostate cancer cells to docetaxel-induced apoptosis. Conversely, overexpression of EZH2 blocked docetaxel-induced apoptosis. We further demonstrated that miR-205 silencing is linked to miR-31 silencing through EZH2. Suppression of miR-205 with an miRNA inhibitor caused an increase of EZH2 protein, which in turn inhibited miR-31 expression. Conversely, overexpression of miR-205 decreased EZH2 protein and increased miR-31 expression. In paired human prostate cancer specimens and adjacent normal tissues, we observed that the decrease of miR-205 expression correlated with EZH2 overexpression and miR-31 silencing. Thus, EZH2 integrates the epigenetic silencing of miR-205 and miR-31 to confer resistance to chemotherapy-induced apoptosis.
- Published
- 2014
- Full Text
- View/download PDF
17. Introduction to manuscript by Prof. Donald S. Coffey.
- Author
-
Tindall DJ
- Published
- 2014
- Full Text
- View/download PDF
18. Modulation of androgen receptor by FOXA1 and FOXO1 factors in prostate cancer.
- Author
-
Zhao Y, Tindall DJ, and Huang H
- Subjects
- Androgens metabolism, Animals, Forkhead Box Protein O1, Humans, Male, Forkhead Transcription Factors metabolism, Hepatocyte Nuclear Factor 3-alpha metabolism, Prostatic Neoplasms metabolism, Receptors, Androgen metabolism
- Abstract
Androgens and the androgen receptor (AR) are essential for growth and differentiation of the normal prostate gland as well as proliferation and survival of prostate cancer (PCa). Increasing evidence suggests that reactivation of the AR plays a pivotal role in disease progression to castration-resistant PCa (CRPC). Forkhead box (FOX) factors exert two distinct effects on AR function in PCa. The A-class of FOX proteins, especially FOXA1, functions as a pioneer factor to facilitate AR transactivation and PCa growth. In contrast, the O-class of FOX proteins such as FOXO1 and FOXO3, which are downstream effectors of the PTEN tumor suppressor, inhibit the transcriptional activity of either full-length AR or constitutively active splice variants of AR in a direct or indirect manner in PCa. FOXO1 also contributes to taxane-mediated inhibition of the AR and CRPC growth. Therefore, FOX family members not only have a tight relationship with AR, but also represent a pivotal group of proteins to be targeted for PCa therapy. The present review focuses primarily on recent advances in the epigenetic, mechanistic and clinical relevant aspects of regulation of the AR by FOXA1 and FOXO1 factors in PCa.
- Published
- 2014
- Full Text
- View/download PDF
19. Preclinical evaluation of the supercritical extract of azadirachta indica (neem) leaves in vitro and in vivo on inhibition of prostate cancer tumor growth.
- Author
-
Wu Q, Kohli M, Bergen HR 3rd, Cheville JC, Karnes RJ, Cao H, Young CY, Tindall DJ, McNiven MA, and Donkena KV
- Subjects
- Animals, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents toxicity, Apoptosis drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Disease Models, Animal, Focal Adhesions drug effects, Humans, Inhibitory Concentration 50, Male, Mice, Plant Extracts pharmacokinetics, Plant Extracts toxicity, Prostate-Specific Antigen metabolism, Prostatic Neoplasms drug therapy, Prostatic Neoplasms metabolism, Receptors, Androgen metabolism, Tumor Burden drug effects, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Azadirachta chemistry, Plant Extracts pharmacology, Plant Leaves chemistry, Prostatic Neoplasms pathology
- Abstract
Azadirachta indica, commonly known as neem, has gained worldwide prominence because of its medical properties, namely antitumor, antiviral, anti-inflammatory, antihyperglycemic, antifungal, and antibacterial activities. Despite these promising results, gaps remain in our understanding of the molecular mechanism of action of neem compounds and their potential for use in clinical trials. We investigated supercritical extract of neem leaves (SENL) for the following: molecular targets in vitro, in vivo efficacy to inhibit tumor growth, and bioactive compounds that exert antitumor activity. Treatment of LNCaP-luc2 prostate cancer cells with SENL suppressed dihydrotestosterone-induced androgen receptor and prostate-specific antigen levels. SENL inhibited integrin β1, calreticulin, and focal adhesion kinase activation in LNCaP-luc2 and PC3 prostate cancer cells. Oral administration of SENL significantly reduced LNCaP-luc2 xenograft tumor growth in mice with the formation of hyalinized fibrous tumor tissue, reduction in the prostate-specific antigen, and increase in AKR1C2 levels. To identify the active anticancer compounds, we fractionated SENL by high-pressure liquid chromatography and evaluated 16 peaks for cytotoxic activity. Four of the 16 peaks exhibited significant cytotoxic activity against prostate cancer cells. Mass spectrometry of the isolated peaks suggested the compounds with cytotoxic activity were nimbandiol, nimbolide, 2',3'-dihydronimbolide, and 28-deoxonimbolide. Analysis of tumor tissue and plasma samples from mice treated with SENL indicated 28-deoxonimbolide and nimbolide as the bioactive compounds. Overall, our data revealed the bioactive compounds in SENL and suggested that the anticancer activity could be mediated through alteration in androgen receptor and calreticulin levels in prostate cancer.
- Published
- 2014
- Full Text
- View/download PDF
20. Analyzing site selectivity in Rh2(esp)2-catalyzed intermolecular C-H amination reactions.
- Author
-
Bess EN, DeLuca RJ, Tindall DJ, Oderinde MS, Roizen JL, Du Bois J, and Sigman MS
- Subjects
- Amination, Carbon chemistry, Catalysis, Hydrogen chemistry, Models, Chemical, Rhodium chemistry
- Abstract
Predicting site selectivity in C-H bond oxidation reactions involving heteroatom transfer is challenged by the small energetic differences between disparate bond types and the subtle interplay of steric and electronic effects that influence reactivity. Herein, the factors governing selective Rh2(esp)2-catalyzed C-H amination of isoamylbenzene derivatives are investigated, where modification to both the nitrogen source, a sulfamate ester, and substrate are shown to impact isomeric product ratios. Linear regression mathematical modeling is used to define a relationship that equates both IR stretching parameters and Hammett σ(+) values to the differential free energy of benzylic versus tertiary C-H amination. This model has informed the development of a novel sulfamate ester, which affords the highest benzylic-to-tertiary site selectivity (9.5:1) observed for this system.
- Published
- 2014
- Full Text
- View/download PDF
21. CBP loss cooperates with PTEN haploinsufficiency to drive prostate cancer: implications for epigenetic therapy.
- Author
-
Ding L, Chen S, Liu P, Pan Y, Zhong J, Regan KM, Wang L, Yu C, Rizzardi A, Cheng L, Zhang J, Schmechel SC, Cheville JC, Van Deursen J, Tindall DJ, and Huang H
- Subjects
- Animals, Cell Line, Tumor, Cyclin-Dependent Kinase Inhibitor p27 genetics, Enhancer of Zeste Homolog 2 Protein, Histones metabolism, Humans, Hydroxamic Acids pharmacology, Indoles pharmacology, Male, Membrane Proteins physiology, Mice, PTEN Phosphohydrolase physiology, Panobinostat, Peptide Fragments genetics, Phosphoproteins physiology, Polycomb Repressive Complex 2 physiology, Prostatic Neoplasms therapy, Proto-Oncogene Proteins c-akt physiology, Sialoglycoproteins genetics, ras GTPase-Activating Proteins genetics, Epigenesis, Genetic, Haploinsufficiency, PTEN Phosphohydrolase genetics, Peptide Fragments physiology, Prostatic Neoplasms genetics, Sialoglycoproteins physiology
- Abstract
Despite the high incidence and mortality of prostate cancer, the etiology of this disease is not fully understood. In this study, we develop functional evidence for CBP and PTEN interaction in prostate cancer based on findings of their correlate expression in the human disease. Cbp(pc-/-);Pten(pc+/-) mice exhibited higher cell proliferation in the prostate and an early onset of high-grade prostatic intraepithelial neoplasia. Levels of EZH2 methyltransferase were increased along with its Thr350 phosphorylation in both mouse Cbp(-/-); Pten(+/-) and human prostate cancer cells. CBP loss and PTEN deficiency cooperated to trigger a switch from K27-acetylated histone H3 to K27-trimethylated bulk histones in a manner associated with decreased expression of the growth inhibitory EZH2 target genes DAB2IP, p27(KIP1), and p21(CIP1). Conversely, treatment with the histone deacetylase inhibitor panobinostat reversed this switch, in a manner associated with tumor suppression in Cbp(pc-/-);Pten(pc+/-) mice. Our findings show how CBP and PTEN interact to mediate tumor suppression in the prostate, establishing a central role for histone modification in the etiology of prostate cancer and providing a rationale for clinical evaluation of epigenetic-targeted therapy in patients with prostate cancer., (©2014 AACR.)
- Published
- 2014
- Full Text
- View/download PDF
22. p300 acetyltransferase regulates androgen receptor degradation and PTEN-deficient prostate tumorigenesis.
- Author
-
Zhong J, Ding L, Bohrer LR, Pan Y, Liu P, Zhang J, Sebo TJ, Karnes RJ, Tindall DJ, van Deursen J, and Huang H
- Subjects
- Aged, Animals, Cell Line, Tumor, Cell Proliferation, Female, HEK293 Cells, Humans, Male, Mice, Mice, Knockout, Middle Aged, PTEN Phosphohydrolase deficiency, Phosphorylation, Prostatic Intraepithelial Neoplasia pathology, Prostatic Neoplasms pathology, Proteolysis, Transcription, Genetic, Ubiquitination, Carcinogenesis metabolism, PTEN Phosphohydrolase genetics, Prostatic Intraepithelial Neoplasia enzymology, Prostatic Neoplasms enzymology, Receptors, Androgen metabolism, p300-CBP Transcription Factors physiology
- Abstract
Overexpression of the histone acetyltransferase p300 is implicated in the proliferation and progression of prostate cancer, but evidence of a causal role is lacking. In this study, we provide genetic evidence that this generic transcriptional coactivator functions as a positive modifier of prostate tumorigenesis. In a mouse model of PTEN deletion-induced prostate cancer, genetic ablation of p300 attenuated expression of the androgen receptor (AR). This finding was confirmed in human prostate cancer cells in which PTEN expression was abolished by RNA interference-mediated attenuation. These results were consistent with clinical evidence that the expression of p300 and AR correlates positively in human prostate cancer specimens. Mechanistically, PTEN inactivation increased AR phosphorylation at serine 81 (Ser81) to promote p300 binding and acetylation of AR, thereby precluding its polyubiquitination and degradation. In support of these findings, in PTEN-deficient prostate cancer in the mouse, we found that p300 was crucial for AR target gene expression. Taken together, our work identifies p300 as a molecular determinant of AR degradation and highlights p300 as a candidate target to manage prostate cancer, especially in cases marked by PTEN loss., (©2014 AACR.)
- Published
- 2014
- Full Text
- View/download PDF
23. Imidazolidin-2-one: pseudosymmetry and twinning.
- Author
-
Taouss C, Jones PG, and Tindall DJ
- Abstract
The title compound, C3H6N2O, crystallizes with imposed twofold symmetry in the space group I4(1)/a. The five-membered ring displays a half-chair conformation. N-H···O hydrogen bonds connect the molecules to form R2(2)(8) rings and thence ribbons parallel to the a and b axes. These intersect via O2H2 rings involving longer H···O contacts. The crystal was merohedrally twinned. Preliminary indications of the higher symmetry space group I4(1)/amd, which would require the ring to be planar, proved to be incorrect. A previous brief report of the structure in Fdd2 is also probably incorrect.
- Published
- 2013
- Full Text
- View/download PDF
24. Posttranslational modification of the androgen receptor in prostate cancer.
- Author
-
van der Steen T, Tindall DJ, and Huang H
- Subjects
- Acetylation, Humans, Male, Methylation, Phosphoric Monoester Hydrolases metabolism, Phosphorylation, Prostatic Neoplasms pathology, Protein Processing, Post-Translational, Protein Structure, Tertiary, Receptors, Androgen chemistry, Receptors, Androgen genetics, Sumoylation, Ubiquitination, Prostatic Neoplasms metabolism, Receptors, Androgen metabolism
- Abstract
The androgen receptor (AR) is important in the development of the prostate by regulating transcription, cellular proliferation, and apoptosis. AR undergoes posttranslational modifications that alter its transcription activity, translocation to the nucleus and stability. The posttranslational modifications that regulate these events are of utmost importance to understand the functional role of AR and its activity. The majority of these modifications occur in the activation function-1 (AF1) region of the AR, which contains the transcriptional activation unit 1 (TAU1) and 5 (TAU5). Identification of the modifications that occur to these regions may increase our understanding of AR activation in prostate cancer and the role of AR in the progression from androgen-dependent to castration-resistant prostate cancer (CRPC). Most of the posttranslational modifications identified to date have been determined using the full-length AR in androgen dependent cells. Further investigations into the role of posttranslational modifications in androgen-independent activation of full-length AR and constitutively active splicing variants are warranted, findings from which may provide new therapeutic options for CRPC.
- Published
- 2013
- Full Text
- View/download PDF
25. Androgens regulate TRAIL-induced cell death in prostate cancer cells via multiple mechanisms.
- Author
-
Wang D, Lu J, and Tindall DJ
- Subjects
- Androstadienes pharmacology, Apoptosis, Cell Cycle drug effects, Cell Line, Tumor, Gene Expression Regulation drug effects, Humans, Male, NF-kappa B metabolism, Nuclear Pore Complex Proteins metabolism, Prostatic Neoplasms, RNA-Binding Proteins metabolism, Receptors, TNF-Related Apoptosis-Inducing Ligand metabolism, Signal Transduction drug effects, TNF Receptor-Associated Death Domain Protein metabolism, Testosterone Congeners physiology, Wortmannin, Antineoplastic Agents pharmacology, Metribolone pharmacology, TNF-Related Apoptosis-Inducing Ligand pharmacology, Testosterone Congeners pharmacology
- Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising therapeutic agent for prostate cancer because it selectively induces apoptosis in cancer cells but not in normal cells. Previous reports have suggested that androgens regulate TRAIL-induced apoptosis in prostate cancer cells. However, there are discrepancies between these reports of how androgens affect TRAIL-induced cell death. To clarify the role of androgens on TRAIL-induced apoptosis in prostate cancer cells, we investigated the effects of androgen on TRAIL-induced cell death in a dose-response manner. Our results showed that although androgens sensitize LNCaP cells to TRAIL-induced apoptosis, this effect is dose-dependent and biphasic. We found that low levels of androgen are superior to high levels of androgen in term of sensitizing LNCaP cells to TRAIL. We also found that upregulation of DR5 (TRAIL-R2) expression by androgens is critical for sensitizing LNCaP cells to TRAIL. However, low levels of androgen are sufficient to induce DR5 expression and sensitize LNCaP cells to TRAIL-induced cell death. High levels of androgen alter the TRADD/RIP1 ratio, which may contribute to NF-κB activation and sequentially inhibit TRAIL-induced apoptosis., (Copyright © 2013 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
26. Androgen receptor: past, present and future.
- Author
-
Schmidt LJ and Tindall DJ
- Subjects
- Androgens genetics, Androgens metabolism, Animals, Genome-Wide Association Study methods, Humans, Male, Mutation, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Signal Transduction, Androgen Antagonists therapeutic use, Prostatic Neoplasms drug therapy, Receptors, Androgen genetics, Receptors, Androgen metabolism
- Abstract
Androgens and the androgen receptor have been the focus of prostate cancer research since the early 1940s, when Huggins and Hodges demonstrated that removal of androgens caused advanced prostate cancer to regress. Since that time, a large number of androgen deprivation therapies have been developed in an effort to cure this disease, but prostate cancer remains one of the leading causes of cancer death in males worldwide. This is due in part to the emergence of castration- recurrent prostate cancer in patients with advanced disease who have failed androgen deprivation therapy. The androgen receptor is still a major player in castration-recurrent disease, and though much has been discovered since the early work of Huggins and Hodges regarding how prostate cancer cells manage to avoid the effects of androgen deprivation, survival times for men with advanced prostate cancer have changed only modestly. Research is now directed toward delineating the mechanisms of action of the androgen receptor under castrate conditions, whether through amplification of the AR, mutation, expression of splice variants, use of alternate signaling pathways, aberrant expression and activation of coregulators, or intratumoral androgen biosynthesis. Genome-wide association studies are also adding to the wealth of knowledge surrounding the androgen receptor, and with this knowledge comes the ability to design new drug therapies directed toward eradication of this disease.
- Published
- 2013
- Full Text
- View/download PDF
27. Autophagy control by the VEGF-C/NRP-2 axis in cancer and its implication for treatment resistance.
- Author
-
Stanton MJ, Dutta S, Zhang H, Polavaram NS, Leontovich AA, Hönscheid P, Sinicrope FA, Tindall DJ, Muders MH, and Datta K
- Subjects
- Apoptosis physiology, Blotting, Western, Cell Line, Tumor, Enzyme-Linked Immunosorbent Assay, Gene Expression Regulation, Neoplastic genetics, Humans, Microscopy, Confocal, Neoplasms genetics, Neuropilin-2 genetics, Oligonucleotide Array Sequence Analysis, Real-Time Polymerase Chain Reaction, Signal Transduction genetics, Transfection, Vascular Endothelial Growth Factor C genetics, Autophagy genetics, Drug Resistance, Neoplasm genetics, Neoplasms metabolism, Neuropilin-2 metabolism, Vascular Endothelial Growth Factor C metabolism
- Abstract
A major contributor to cancer mortality is recurrence and subsequent metastatic transformation following therapeutic intervention. Therefore, in order to develop new treatment modalities and improve the efficacy of current ones, it is important to understand the molecular mechanisms that promote resistance to therapy in cancer cells. One pathway contributing to therapy resistance is autophagy, a self-digestive process that can eliminate unnecessary or damaged organelles to protect cancer cells from death. We have found that the VEGF-C/NRP-2 axis is involved in the activation of autophagy, which helps cancer cell survival following treatment. Inhibition of mTOR complex 1 activity by this axis is the underlying mechanism for the activation of autophagy. Furthermore, we identified two VEGF-C/NRP-2-regulated genes, LAMP-2 and WDFY-1, that have previously been suggested to participate in autophagy and vesicular trafficking. Upregulation of WDFY-1 following VEGF-C or NRP-2 depletion contributes to cytotoxic drug-mediated cell death. Together, these data suggest a link between the VEGF-C/NRP-2 axis and cancer cell survival despite the presence of chemotherapy-induced stress. Effective targeting of this pathway may lead to the development of new cancer therapies.
- Published
- 2013
- Full Text
- View/download PDF
28. Global methylation profiling for risk prediction of prostate cancer.
- Author
-
Mahapatra S, Klee EW, Young CY, Sun Z, Jimenez RE, Klee GG, Tindall DJ, and Donkena KV
- Subjects
- Biomarkers, Tumor genetics, DNA, Neoplasm genetics, DNA, Neoplasm metabolism, Humans, Male, Middle Aged, Neoplasm Recurrence, Local genetics, Prognosis, Promoter Regions, Genetic, Prostatic Neoplasms diagnosis, DNA Methylation, DNA, Neoplasm chemistry, Neoplasm Recurrence, Local diagnosis, Prostatic Neoplasms genetics
- Abstract
Purpose: The aim of this study was to investigate the promoter hypermethylation as diagnostic markers to detect malignant prostate cells and as prognostic markers to predict the clinical recurrence of prostate cancer., Experimental Design: DNA was isolated from prostate cancer and normal adjacent tissues. After bisulfite conversion, methylation of 14,495 genes was evaluated using the Methylation27 microarrays in 238 prostate tissues. We analyzed methylation profiles in four different groups: (i) tumor (n = 198) versus matched normal tissues (n = 40), (ii) recurrence (n = 123) versus nonrecurrence (n = 75), (iii) clinical recurrence (n = 80) versus biochemical recurrence (n = 43), and (iv) systemic recurrence (n = 36) versus local recurrence (n = 44). Group 1, 2, 3, and 4 genes signifying biomarkers for diagnosis, prediction of recurrence, clinical recurrence, and systemic progression were determined. Univariate and multivariate analyses were conducted to predict risk of recurrence. We validated the methylation of genes in 20 independent tissues representing each group by pyrosequencing., Results: Microarray analysis revealed significant methylation of genes in four different groups of prostate cancer tissues. The sensitivity and specificity of methylation for 25 genes from 1, 2, and 4 groups and 7 from group 3 were shown. Validation of genes by pyrosequencing from group 1 (GSTP1, HIF3A, HAAO, and RARβ), group 2 (CRIP1, FLNC, RASGRF2, RUNX3, and HS3ST2), group 3 (PHLDA3, RASGRF2, and TNFRSF10D), and group 4 (BCL11B, POU3F3, and RASGRF2) confirmed the microarray results., Conclusions: Our study provides a global assessment of DNA methylation in prostate cancer and identifies the significance of genes as diagnostic and progression biomarkers of prostate cancer., (©2012 AACR.)
- Published
- 2012
- Full Text
- View/download PDF
29. RhoA as a mediator of clinically relevant androgen action in prostate cancer cells.
- Author
-
Schmidt LJ, Duncan K, Yadav N, Regan KM, Verone AR, Lohse CM, Pop EA, Attwood K, Wilding G, Mohler JL, Sebo TJ, Tindall DJ, and Heemers HV
- Subjects
- Androgens adverse effects, Animals, Cell Nucleus drug effects, Cell Nucleus metabolism, Cell Proliferation drug effects, DNA-Binding Proteins antagonists & inhibitors, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Humans, LIM-Homeodomain Proteins genetics, LIM-Homeodomain Proteins metabolism, Male, Mice, Mice, Nude, Muscle Proteins genetics, Muscle Proteins metabolism, Neoplasm Proteins agonists, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins genetics, Neoplasm Recurrence, Local, Oncogene Proteins, Fusion antagonists & inhibitors, Oncogene Proteins, Fusion genetics, Oncogene Proteins, Fusion metabolism, Promoter Regions, Genetic drug effects, Prostate drug effects, Prostate metabolism, Prostate pathology, Prostatic Neoplasms pathology, Prostatic Neoplasms surgery, Protein Kinase Inhibitors pharmacology, Protein Transport drug effects, Receptors, Androgen genetics, Recombinant Proteins agonists, Recombinant Proteins antagonists & inhibitors, Recombinant Proteins metabolism, Trans-Activators, Transcription Factors genetics, Transcription Factors metabolism, rho-Associated Kinases antagonists & inhibitors, rho-Associated Kinases genetics, rho-Associated Kinases metabolism, rhoA GTP-Binding Protein agonists, rhoA GTP-Binding Protein antagonists & inhibitors, rhoA GTP-Binding Protein genetics, Androgens pharmacology, Gene Expression Regulation, Neoplastic drug effects, Neoplasm Proteins metabolism, Prostatic Neoplasms metabolism, Receptors, Androgen metabolism, Signal Transduction drug effects, rhoA GTP-Binding Protein metabolism
- Abstract
Recently, we have identified serum response factor (SRF) as a mediator of clinically relevant androgen receptor (AR) action in prostate cancer (PCa). Genes that rely on SRF for androgen responsiveness represent a small fraction of androgen-regulated genes, but distinguish benign from malignant prostate, correlate with aggressive disease, and are associated with biochemical recurrence. Thus, understanding the mechanism(s) by which SRF conveys androgen regulation to its target genes may provide novel opportunities to target clinically relevant androgen signaling. Here, we show that the small GTPase ras homolog family member A (RhoA) mediates androgen-responsiveness of more than half of SRF target genes. Interference with expression of RhoA, activity of the RhoA effector Rho-associated coiled-coil containing protein kinase 1 (ROCK), and actin polymerization necessary for nuclear translocation of the SRF cofactor megakaryocytic acute leukemia (MAL) prevented full androgen regulation of SRF target genes. Androgen treatment induced RhoA activation, increased the nuclear content of MAL, and led to MAL recruitment to the promoter of the SRF target gene FHL2. In clinical specimens RhoA expression was higher in PCa cells than benign prostate cells, and elevated RhoA expression levels were associated with aggressive disease features and decreased disease-free survival after radical prostatectomy. Overexpression of RhoA markedly increased the androgen-responsiveness of select SRF target genes, in a manner that depends on its GTPase activity. The use of isogenic cell lines and a xenograft model that mimics the transition from androgen-stimulated to castration-recurrent PCa indicated that RhoA levels are not altered during disease progression, suggesting that RhoA expression levels in the primary tumor determine disease aggressiveness. Androgen-responsiveness of SRF target genes in castration-recurrent PCa cells continued to rely on AR, RhoA, SRF, and MAL and the presence of intact SRF binding sites. Silencing of RhoA, use of Rho-associated coiled-coil containing protein kinase 1 inhibitors, or an inhibitor of SRF-MAL interaction attenuated (androgen-regulated) cell viability and blunted PCa cell migration. Taken together, these studies demonstrate that the RhoA signaling axis mediates clinically relevant AR action in PCa.
- Published
- 2012
- Full Text
- View/download PDF
30. Dihydrotestosterone synthesis from adrenal precursors does not involve testosterone in castration-resistant prostate cancer.
- Author
-
Campbell TJ, Tindall DJ, and Figg WD
- Subjects
- Disease Progression, Humans, Male, Molecular Targeted Therapy, Neoplasms, Hormone-Dependent drug therapy, Neoplasms, Hormone-Dependent metabolism, Neoplasms, Hormone-Dependent pathology, Neoplasms, Hormone-Dependent surgery, Orchiectomy, Prostatic Neoplasms drug therapy, Prostatic Neoplasms pathology, Prostatic Neoplasms surgery, Testosterone metabolism, Dihydrotestosterone metabolism, Prostatic Neoplasms metabolism, Receptors, Androgen metabolism
- Abstract
Androgen deprivation therapy is the frontline treatment for metastatic prostate cancer; however, because the majority of cases of advanced prostate cancer progress to castration-resistant prostate cancer (CRPC), there is a considerable need to better understand the synthesis of intratumoral concentrations of the androgen receptor (AR) agonist, 5α-dihydrotestosterone (DHT) in CRPC. In a recent article in the Proceedings of the National Academy of Sciences, Chang et al. show that, contrary to widely held assumptions, the dominant pathway to DHT synthesis does not involve testosterone as a precursor to DHT, but instead involves the conversion of Δ ( 4) -androstenedione (AD) to 5α-dione (AD→5α-dione→DHT) by the steroid-5α-reductase isoenzyme 1 (SRD5A1). The authors show that it is this alternative pathway that drives the progression of CRPC, and they confirm these findings in six established human prostate cancer cell lines as well as in the metastatic tumors from two patients with CRPC. Such findings open the door to new areas of research and to the development of new therapeutic targets in CRPC.
- Published
- 2012
- Full Text
- View/download PDF
31. Androgens repress expression of the F-box protein Skp2 via p107 dependent and independent mechanisms in LNCaP prostate cancer cells.
- Author
-
Jiang J, Pan Y, Regan KM, Wu C, Zhang X, Tindall DJ, and Huang H
- Subjects
- Blotting, Northern, Blotting, Western, Cell Line, Tumor, Cell Proliferation drug effects, Flow Cytometry, Humans, Male, Metribolone pharmacology, Prostatic Neoplasms enzymology, Prostatic Neoplasms genetics, S-Phase Kinase-Associated Proteins genetics, S-Phase Kinase-Associated Proteins metabolism, Testosterone Congeners pharmacology, Cell Cycle physiology, Prostatic Neoplasms metabolism, Receptors, Androgen metabolism, Retinoblastoma-Like Protein p107 metabolism, S-Phase Kinase-Associated Proteins biosynthesis
- Abstract
Background: Androgens control homeostasis of the normal prostate and growth of prostate cancer (PCa) through the androgen receptor (AR) by regulating gene networks involving in cell proliferation, differentiation, and survival. We demonstrated previously that expression of Skp2, a key protein regulating cell entry into the S phase, is inhibited by androgens in an AR-dependent manner (Oncogene, 2004; 23(12): 2161-2176). However, the underlying mechanism of this regulation is unknown., Methods: Using the LNCaP PCa cell line as a working model, the effect of androgens on the expression of Skp2 was examined by Western and Northern blot analyses. Cell cycle was measured by fluorescence-activated cell sorting (FACS). Gene transfection was performed by electroporation to manipulate the expression levels of proteins studied., Results: At physiological levels androgens markedly repressed Skp2 expression but slightly induced Skp2 expression at subphysiological levels. Androgens modestly decreased the stability of the Skp2 protein. Androgenic repression of Skp2 expression was completely abolished by E1A-mediated inactivation of pocket proteins including RB, p130, and p107. Moreover, ectopic expression of p107 inhibited Skp2 expression, and silencing of p107 partially blocked androgenic repression of Skp2., Conclusions: Our data indicate that androgens repress Skp2 expression via p107-dependent and -independent pathways in PCa cells. These regulatory mechanisms may be targeted for the development of new therapeutics of androgen-refractory PCa., (Copyright © 2011 Wiley Periodicals, Inc.)
- Published
- 2012
- Full Text
- View/download PDF
32. Use of tumor dynamics to clarify the observed variability among biochemical recurrence nomograms for prostate cancer.
- Author
-
Dimonte G, Bergstralh EJ, Bolander ME, Karnes RJ, and Tindall DJ
- Subjects
- Algorithms, Cell Proliferation, Cohort Studies, Humans, Male, Prognosis, Prostate-Specific Antigen blood, Prostatectomy, Prostatic Neoplasms surgery, Retrospective Studies, Risk Factors, Seminal Vesicles pathology, Disease Progression, Models, Biological, Neoplasm Recurrence, Local epidemiology, Nomograms, Prostatic Neoplasms diagnosis, Prostatic Neoplasms pathology
- Abstract
Background: Nomograms for biochemical recurrence (BCR) of prostate cancer (PC) after radical prostatectomy can yield very different prognoses for individual patients. Since the nomograms are optimized on different cohorts, the variations may be due to differences in patient risk-factor distributions. In addition, the nomograms assign different relative scores to the same PC risk factors and rarely stratify for tumor growth rate., Methods: We compared BCR-free probabilities from the GPSM model with a cell kinetics (CK) model that uses the individual's tumor state and growth rate. We first created a cohort of 143 patients that reproduced the GPSM patient distribution in Gleason score, Prostate specific antigen (PSA), Seminal vesicle involvement and Margin status since they form the GPSM score. We then performed 143 CK calculations to determine BCR-free probabilities for comparison with the GPSM results for all scores and with four other prominent nomograms for a high-risk patient., Results: The BCR-free probabilities from the CK model agree within 10% with those from the GPSM study for all scores once the CK model parameters are stratified in terms of the GPSM risk factors and the PSA doubling time (PSADT). However, the probabilities from widely used nomograms vary significantly., Conclusions: The CK model reproduces the observed GPSM BCR-free probabilities with a broad stratification of model parameters for PC risk factors and can thus be used to describe PC progression for individual patients. The analysis suggests that nomograms should stratify for PSADT to be predictive., (Copyright © 2011 Wiley Periodicals, Inc.)
- Published
- 2012
- Full Text
- View/download PDF
33. Antiangiogenic Effects and Therapeutic Targets of Azadirachta indica Leaf Extract in Endothelial Cells.
- Author
-
Mahapatra S, Young CY, Kohli M, Karnes RJ, Klee EW, Holmes MW, Tindall DJ, and Donkena KV
- Abstract
Azadirachta indica (common name: neem) leaves have been found to possess immunomodulatory, anti-inflammatory and anti-carcinogenic properties. The present study evaluates anti-angiogenic potential of ethanol extract of neem leaves (EENL) in human umbilical vein endothelial cells (HUVECs). Treatment of HUVECs with EENL inhibited VEGF induced angiogenic response in vitro and in vivo. The in vitro proliferation, invasion and migration of HUVECs were suppressed with EENL. Nuclear fragmentation and abnormally small mitochondria with dilated cristae were observed in EENL treated HUVECs by transmission electron microscopy. Genome-wide mRNA expression profiling after treatment with EENL revealed differentially regulated genes. Expression changes of the genes were validated by quantitative real-time polymerase chain reaction. Additionally, increase in the expression of HMOX1, ATF3 and EGR1 proteins were determined by immunoblotting. Analysis of the compounds in the EENL by mass spectrometry suggests the presence of nimbolide, 2',3'-dehydrosalannol, 6-desacetyl nimbinene and nimolinone. We further confirmed antiproliferative activity of nimbolide and 2',3'-dehydrosalannol in HUVECs. Our results suggest that EENL by regulating the genes involved in cellular development and cell death functions could control cell proliferation, attenuate the stimulatory effects of VEGF and exert antiangiogenic effects. EENL treatment could have a potential therapeutic role during cancer progression.
- Published
- 2012
- Full Text
- View/download PDF
34. Regulation of FOXO protein stability via ubiquitination and proteasome degradation.
- Author
-
Huang H and Tindall DJ
- Subjects
- Animals, Forkhead Box Protein O1, Humans, Models, Biological, Protein Binding, Forkhead Transcription Factors metabolism, Proto-Oncogene Proteins c-akt metabolism, S-Phase Kinase-Associated Proteins metabolism, Ubiquitination
- Abstract
Forkhead box O-class (FOXO) proteins are evolutionally conserved transcription factors. They belong to a family of proteins consisting of FOXO1, FOXO3a, FOXO4 and FOXO6 in humans. Increasing evidence suggests that FOXO proteins function as tumor suppressors by transcriptionally regulating expression of genes involved in cell cycle arrest, apoptosis, DNA repair and oxidative stress resistance. Activation of various protein kinases, including Akt, IκB kinase (IKK) and ERK, leads to phosphorylation of FOXO proteins and their ubiquitination mediated by E3 ligases such as SKP2 and MDM2 in human primary tumors and cancer cell lines. As a result, the tumor suppressor functions of FOXO proteins are either diminished or abrogated due to their ubiquitination-proteasome degradation, thereby favoring cell transformation, proliferation and survival. Thus, ubiquitination and proteasome degradation of FOXO proteins play an important role in tumorigenesis and represent a viable target for cancer treatment. This article is part of a Special Issue entitled: PI3K-AKT-FoxO axis in cancer and aging., (2011 Elsevier B.V. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
35. Alternatively spliced androgen receptor variants.
- Author
-
Dehm SM and Tindall DJ
- Subjects
- Androgen-Insensitivity Syndrome metabolism, Animals, Exons, Gene Expression Regulation, Neoplastic, Humans, Male, Prostatic Neoplasms metabolism, Protein Isoforms, Receptors, Androgen metabolism, Alternative Splicing, Androgen-Insensitivity Syndrome genetics, Prostatic Neoplasms genetics, Receptors, Androgen genetics
- Abstract
Alternative splicing is an important mechanism for increasing functional diversity from a limited set of genes. Deregulation of this process is common in diverse pathologic conditions. The androgen receptor (AR) is a steroid receptor transcription factor with functions critical for normal male development as well as the growth and survival of normal and cancerous prostate tissue. Studies of AR function in androgen insensitivity syndrome (AIS) and prostate cancer (PCa) have demonstrated loss-of-function AR alterations in AIS and gain-of-function AR alterations in PCa. Over the past two decades, AR gene alterations have been identified in various individuals with AIS, which disrupt normal AR splicing patterns and yield dysfunctional AR protein variants. Recently, altered AR splicing patterns have been identified as a mechanism of PCa progression and resistance to androgen depletion therapy. Several studies have described the synthesis of alternatively spliced transcripts encoding truncated AR isoforms that lack the ligand-binding domain, which is the ultimate target of androgen depletion. Many of these truncated AR isoforms function as constitutively active, ligand-independent transcription factors that can support androgen-independent expression of AR target genes, as well as the androgen-independent growth of PCa cells. In this review, we will summarize the various alternatively spliced AR variants that have been discovered, with a focus on their role and origin in the pathologic conditions of AIS and PCa.
- Published
- 2011
- Full Text
- View/download PDF
36. Androgen receptor rediscovered: the new biology and targeting the androgen receptor therapeutically.
- Author
-
Ryan CJ and Tindall DJ
- Subjects
- Androgens biosynthesis, Castration, Drug Delivery Systems, Humans, Ligands, Male, Prostatic Neoplasms physiopathology, Signal Transduction, Prostatic Neoplasms drug therapy, Receptors, Androgen physiology
- Abstract
Discoveries over the past decade suggest that castration-resistant prostate cancer (CRPC) is sensitive, but not resistant to, further manipulation of the androgen-androgen receptor (AR) axis. Several new therapies that target this axis have demonstrated clinical activity. In this article, preclinical and clinical findings occurring in the field of AR-targeted therapies are reviewed. Reviews of scientific and clinical development are divided into those occurring prereceptor (androgen production and conversion) and at the level of the receptor (AR aberrations and therapies targeting AR directly). Intracrine androgen production and AR amplification, among others, are among the principal aberrancies driving CRPC growth. Phase III data with abiraterone acetate and phase II data with MDV-3100, along with other similar therapies, confirm for the clinician that the scientific findings related to persistent AR signaling in a castrate milieu can be harnessed to produce significant clinical benefit for patients with the disease. Studies aimed at optimizing the timing of their use and exploring the mechanisms of resistance to these therapies are under way. The clinical success of therapies that directly target androgen synthesis as well as the most common aberrancies of the AR confirm that prostate cancer retains dependence on AR signaling, even in the castrate state.
- Published
- 2011
- Full Text
- View/download PDF
37. Novel molecular targets of Azadirachta indica associated with inhibition of tumor growth in prostate cancer.
- Author
-
Mahapatra S, Karnes RJ, Holmes MW, Young CY, Cheville JC, Kohli M, Klee EW, Tindall DJ, and Donkena KV
- Subjects
- Animals, Antineoplastic Agents, Phytogenic isolation & purification, Blotting, Western, Gene Expression Profiling, Genome-Wide Association Study, Humans, Male, Mice, Mice, Nude, Plant Extracts isolation & purification, Plant Extracts therapeutic use, Plant Leaves chemistry, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Real-Time Polymerase Chain Reaction, Xenograft Model Antitumor Assays, Antineoplastic Agents, Phytogenic therapeutic use, Apoptosis drug effects, Azadirachta chemistry, Cell Proliferation drug effects, Gene Expression drug effects, Prostatic Neoplasms drug therapy
- Abstract
Advanced prostate cancer has significant long-term morbidity, and there is a growing interest in alternative and complimentary forms of therapy that will improve the outcomes of patients. Azadirachta indica (common name: neem) contains multiple active compounds that have potent anti-inflammatory and anticancer properties. The present study investigates the novel targets of the anticancer activity of ethanol extract of neem leaves (EENL) in vitro and evaluates the in vivo efficacy in the prostate cancer models. Analysis of the components in the EENL by mass spectrometry suggests the presence of 2',3'-dehydrosalannol, 6-desacetyl nimbinene, and nimolinone. Treatment of C4-2B and PC-3M-luc2 prostate cancer cells with EENL inhibited the cell proliferation. Genome-wide expression profiling, using oligonucleotide microarrays, revealed genes differentially expressed with EENL treatment in prostate cancer cells. Functional analysis unveiled that most of the up-regulated genes were associated with cell death, and drug metabolism, and the down-regulated genes were associated with cell cycle, DNA replication, recombination, and repair functions. Quantitative PCR confirmed significant up-regulation of 40 genes and immunoblotting revealed increase in the protein expression levels of HMOX1, AKR1C2, AKR1C3, and AKR1B10. EENL treatment inhibited the growth of C4-2B and PC-3M-luc2 prostate cancer xenografts in nude mice. The suppression of tumor growth is associated with the formation of hyalinized fibrous tumor tissue and the induction of cell death by apoptosis. These results suggest that EENL-containing natural bioactive compounds could have potent anticancer property and the regulation of multiple cellular pathways could exert pleiotrophic effects in prevention and treatment of prostate cancer.
- Published
- 2011
- Full Text
- View/download PDF
38. Minireview: Alternative activation pathways for the androgen receptor in prostate cancer.
- Author
-
Lamont KR and Tindall DJ
- Subjects
- Animals, Cytokines metabolism, Gene Expression Regulation, Neoplastic, Humans, Intercellular Signaling Peptides and Proteins metabolism, Male, Mutation, Prostatic Neoplasms genetics, Prostatic Neoplasms physiopathology, Protein Kinases metabolism, Receptors, Androgen genetics, Prostatic Neoplasms metabolism, Receptors, Androgen metabolism, Signal Transduction genetics
- Abstract
Advanced prostate tumors, which are androgen dependent, are often initially treated in the clinic with hormone ablation therapy, either through surgical castration or administration of small-molecule antiandrogens. Most tumors respond favorably to these treatments, exhibiting regression of the tumor, amelioration of symptoms, and a decrease of prostate-specific antigen in patient sera. However, with time, the majority of tumors recur in a more aggressive, castration-resistant (CR) phenotype. Currently, no effective treatment exists for this stage of the cancer, and patients ultimately succumb to metastatic disease. The androgen receptor (AR), which is a member of the nuclear hormone receptor superfamily of proteins, is the transcription factor that is responsible for mediating the effects of androgens upon target tissues, and it has been demonstrated to play a central role in the development and progression of prostate cancer. Despite CR tumor cells being able to continue to grow after hormonal therapy in which testosterone and dihydrotestosterone are markedly reduced, they still require the expression and activity of the AR. The AR can become transactivated in this low-androgen environment through a number of different mechanisms, including amplification and mutation of the receptor, cross talk with other signaling pathways, and altered regulation by coregulatory proteins. This review will summarize the most current data regarding non-ligand-mediated activation of the AR in prostate cancer cells. Developing work in this field aims to more clearly elucidate the signals that drive AR activity independently of androgens in CR disease so that better therapeutic targets can be developed for patients with this stage of highly aggressive prostate carcinoma.
- Published
- 2011
- Full Text
- View/download PDF
39. Targeting 5α-reductase for prostate cancer prevention and treatment.
- Author
-
Nacusi LP and Tindall DJ
- Subjects
- Animals, Dihydrotestosterone metabolism, Humans, Male, Prostatic Neoplasms drug therapy, Signal Transduction drug effects, Signal Transduction physiology, Testosterone metabolism, Treatment Outcome, 3-Oxo-5-alpha-Steroid 4-Dehydrogenase metabolism, 5-alpha Reductase Inhibitors therapeutic use, Drug Delivery Systems methods, Prostatic Neoplasms enzymology, Prostatic Neoplasms prevention & control
- Abstract
Testosterone is the most abundant circulating androgen, and can be converted to dihydrotestosterone (DHT), a more potent androgen, by the 5α-reductase enzymes in target tissues. Current treatments for prostate cancer consist of reducing androgen levels by chemical or surgical castration or pure antiandrogen therapy that directly targets the androgen receptor (AR). Although these therapies reduce tumor burden and AR activity, the cancer inevitably recurs within 18-30 months. An approach targeting the androgen-AR axis at different levels could, therefore, improve the efficacy of prostate cancer therapy. Inhibition of 5α-reductase is one such approach; however, the two largest trials to investigate the use of the 5α-reductase inhibitors (5ARIs) finasteride and dutasteride in patients with prostate cancer have shown that, although the incidence of cancer was reduced by 5ARI treatment, those cancers that were detected were more aggressive than in patients treated with placebo. Thus, the best practice for using these drugs to prevent and treat prostate cancer remains unclear.
- Published
- 2011
- Full Text
- View/download PDF
40. Steroid 5 α-reductase inhibitors targeting BPH and prostate cancer.
- Author
-
Schmidt LJ and Tindall DJ
- Subjects
- 5-alpha Reductase Inhibitors chemistry, Azasteroids chemistry, Azasteroids therapeutic use, Dutasteride, Finasteride chemistry, Finasteride therapeutic use, Humans, Male, Molecular Structure, 5-alpha Reductase Inhibitors therapeutic use, Clinical Trials as Topic, Prostatic Hyperplasia drug therapy, Prostatic Neoplasms drug therapy
- Abstract
Steroid 5 alpha-reductase inhibitors (5ARIs) have been approved for use clinically in treatment of benign prostate hyperplasia (BPH) and accompanying lower urinary tract symptoms (LUTS) and have also been evaluated in clinical trials for prevention and treatment of prostate cancer. There are currently two steroidal inhibitors in use, finasteride and dutasteride, both with distinct pharmacokinetic properties. This review will examine the evidence presented by various studies supporting the use of these steroidal inhibitors in the prevention and treatment of prostate disease. Article from the Special issue on Targeted Inhibitors., (Copyright © 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
41. Identification of a clinically relevant androgen-dependent gene signature in prostate cancer.
- Author
-
Heemers HV, Schmidt LJ, Sun Z, Regan KM, Anderson SK, Duncan K, Wang D, Liu S, Ballman KV, and Tindall DJ
- Subjects
- Humans, Lasers, Male, Microdissection, Neoplasm Staging, Oligonucleotide Array Sequence Analysis, Prostatic Neoplasms pathology, RNA, Small Interfering, Reverse Transcriptase Polymerase Chain Reaction, Transfection, Biomarkers, Tumor genetics, Gene Expression Profiling, Prostatic Neoplasms genetics, Receptors, Androgen genetics, Serum Response Factor genetics
- Abstract
The androgen receptor (AR) is the principal target for treatment of non-organ-confined prostate cancer (PCa). Androgen deprivation therapies (ADT) directed against the AR ligand-binding domain do not fully inhibit androgen-dependent signaling critical for PCa progression. Thus, information that could direct the development of more effective ADTs is desired. Systems and bioinformatics approaches suggest that considerable variation exists in the mechanisms by which AR regulates expression of effector genes, pointing to a role for secondary transcription factors. A combination of microarray and in silico analyses led us to identify a 158-gene signature that relies on AR along with the transcription factor SRF (serum response factor), representing less than 6% of androgen-dependent genes. This AR-SRF signature is sufficient to distinguish microdissected benign and malignant prostate samples, and it correlates with the presence of aggressive disease and poor outcome. The AR-SRF signature described here associates more strongly with biochemical failure than other AR target gene signatures of similar size. Furthermore, it is enriched in malignant versus benign prostate tissues, compared with other signatures. To our knowledge, this profile represents the first demonstration of a distinct mechanism of androgen action with clinical relevance in PCa, offering a possible rationale to develop novel and more effective forms of ADT., (©2011 AACR.)
- Published
- 2011
- Full Text
- View/download PDF
42. Androgen action during prostate carcinogenesis.
- Author
-
Wang D and Tindall DJ
- Subjects
- Androgens genetics, Animals, Apoptosis genetics, Cell Proliferation, Cell Survival genetics, Cell Transformation, Neoplastic metabolism, Gene Expression Regulation, Neoplastic, Humans, Male, Neoplasm Metastasis, Prostate pathology, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Signal Transduction genetics, Androgens metabolism, Cell Transformation, Neoplastic genetics, Prostate metabolism, Prostatic Neoplasms genetics
- Abstract
Androgens are critical for normal prostate development and function, as well as prostate cancer initiation and progression. Androgens function mainly by regulating target gene expression through the androgen receptor (AR). Many studies have shown that androgen-AR signaling exerts actions on key events during prostate carcinogenesis. In this review, androgen action in distinct aspects of prostate carcinogenesis, including (i) cell proliferation, (ii) cell apoptosis, and (iii) prostate cancer metastasis will be discussed.
- Published
- 2011
- Full Text
- View/download PDF
43. Androgen receptor signaling in prostate cancer development and progression.
- Author
-
Lonergan PE and Tindall DJ
- Abstract
The androgen receptor (AR) signaling axis plays a critical role in the development, function and homeostasis of the prostate. The classical action of AR is to regulate gene transcriptional processes via AR nuclear translocation, binding to androgen response elements on target genes and recruitment of, or crosstalk with, transcription factors. Prostate cancer initiation and progression is also uniquely dependent on AR. Androgen deprivation therapy remains the standard of care for treatment of advanced prostate cancer. Despite an initial favorable response, almost all patients invariably progress to a more aggressive, castrate-resistant phenotype. Considerable evidence now supports the concept that development of castrate-resistant prostate cancer (CRPC) is causally related to continued transactivation of AR. Understanding the critical events and complexities of AR signaling in the progression to CRPC is essential in developing successful future therapies. This review provides a synopsis of AR structure and signaling in prostate cancer progression, with a special focus on recent findings on the role of AR in CRPC. Clinical implications of these findings and potential directions for future research are also outlined.
- Published
- 2011
- Full Text
- View/download PDF
44. Androgen action in prostate cancer.
- Author
-
Basu S and Tindall DJ
- Subjects
- Animals, Humans, Male, Androgens metabolism, Prostatic Neoplasms metabolism
- Abstract
Prostate cancer represents a major health problem in men worldwide. Androgens are required for the growth and maintenance of the prostate. Androgens act by binding to the androgen receptor (AR), a nuclear receptor transcription factor present in the prostate tissues. Most prostate tumors also retain their androgen dependence; therefore, androgen ablation is usually the preferred initial therapeutic approach for the treatment of advanced prostate cancer patients. This review summarizes the current information regarding the role of androgens in prostate cancer., (© Springer Science+Business Media, LLC 2010)
- Published
- 2010
- Full Text
- View/download PDF
45. Differential regulation of steroid nuclear receptor coregulator expression between normal and neoplastic prostate epithelial cells.
- Author
-
Heemers HV, Schmidt LJ, Kidd E, Raclaw KA, Regan KM, and Tindall DJ
- Subjects
- Animals, Blotting, Western, Cell Fractionation, Cell Line, Tumor, Cell Nucleus genetics, Cell Nucleus metabolism, Cells, Cultured, Male, Nuclear Receptor Coactivators metabolism, Prostatic Neoplasms metabolism, Rats, Rats, Wistar, Receptors, Androgen metabolism, Reverse Transcriptase Polymerase Chain Reaction, p300-CBP Transcription Factors metabolism, Nuclear Receptor Coactivators genetics, Prostate metabolism, Prostatic Neoplasms genetics, Receptors, Androgen genetics, p300-CBP Transcription Factors genetics
- Abstract
Background: Deregulated androgen receptor (AR) action is critical for prostate cancer (PCa) progression. Aberrant expression of AR-associated coregulators contributes to AR activity in PCa. The mechanisms underlying coregulator expression in PCa are under intense investigation as they may lead to alternative means of targeting AR activity in PCa cells. We have recently shown that over 30% of coregulator expression in the PCa cell line LNCaP is subject to androgen regulation., Methods: Using multiple PCa cell lines as well as xenograft models, non-malignant prostate epithelial cell lines and androgen-responsive tissues derived from a male Wistar rat model system, we explored the effect of androgen stimulation and androgen deprivation on the expression of the core coactivators SRC1, SRC2, SRC3, CBP, and p300., Results: Androgen stimulation of model systems representing PCa led to a decrease in the expression of SRC1, SRC2, SRC3, CBP, and p300, whereas androgen deprivation induced the expression of these coactivators. In contrast, expression of these coregulators remained largely unaffected following changes in the androgenic milieu in AR-positive models representing non-malignant prostate cells and tissues., Conclusions: Our data indicate differences in the regulation of coregulator expression between neoplastic and normal prostate cells. These findings emphasize the important potential of targeting the mechanisms regulating coregulator expression for therapeutic intervention in PCa.
- Published
- 2010
- Full Text
- View/download PDF
46. FAVL elevation in human tumors disrupts Fanconi anemia pathway signaling and promotes genomic instability and tumor growth.
- Author
-
Zhang J, Zhao D, Park HK, Wang H, Dyer RB, Liu W, Klee GG, McNiven MA, Tindall DJ, Molina JR, and Fei P
- Subjects
- Animals, Base Sequence, Chromosome Aberrations, Disease Progression, Genomic Instability, Humans, Male, Mice, Mice, Nude, Models, Genetic, Molecular Sequence Data, Neoplasm Transplantation, Sequence Homology, Nucleic Acid, Signal Transduction, Carcinoma embryology, Fanconi Anemia metabolism, Fanconi Anemia Complementation Group L Protein metabolism, Gene Expression Regulation, Neoplastic
- Abstract
Fanconi anemia (FA) is a rare human genetic disease caused by mutations in any one of 13 known genes that encode proteins functioning in one common signaling pathway, the FA pathway, or in unknown genes. One characteristic of FA is an extremely high incidence of cancer, indicating the importance of the FA pathway in tumor suppression. However, the role of this pathway in the development and progression of human cancers in individuals who do not have FA has not been clearly determined. Here, we report that elevated expression of what we believe to be a novel splice variant of FA complementation group L (FANCL), which we identified and named FAVL, can impair the FA pathway in non-FA human tumor cells and act as a tumor promoting factor. FAVL expression was elevated in half of the human carcinoma cell lines and carcinoma tissue samples tested. Expression of FAVL resulted in decreased FANCL expression by sequestering FANCL to the cytoplasm and enhancing its degradation. Importantly, this impairment of the FA pathway by FAVL elevation provided human cancer cells with a growth advantage, caused chromosomal instability in vitro, and promoted tumor development in a xenograft mouse model. These data indicate that FAVL impairment of the FA pathway likely contributes to the development of non-FA human cancers and therefore add a challenging layer of complexity to the pathogenesis of human cancer. We further believe that these data will prove useful for developing additional tools for fighting human cancer.
- Published
- 2010
- Full Text
- View/download PDF
47. Effect of dutasteride on the risk of prostate cancer.
- Author
-
Andriole GL, Bostwick DG, Brawley OW, Gomella LG, Marberger M, Montorsi F, Pettaway CA, Tammela TL, Teloken C, Tindall DJ, Somerville MC, Wilson TH, Fowler IL, and Rittmaster RS
- Subjects
- Aged, Azasteroids adverse effects, Biopsy, Double-Blind Method, Dutasteride, Enzyme Inhibitors adverse effects, Erectile Dysfunction chemically induced, Heart Failure chemically induced, Humans, Male, Middle Aged, Prostate pathology, Prostate-Specific Antigen blood, Prostatic Hyperplasia pathology, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Protein Isoforms, Risk, Treatment Outcome, 5-alpha Reductase Inhibitors, Azasteroids therapeutic use, Enzyme Inhibitors therapeutic use, Prostatic Hyperplasia drug therapy, Prostatic Neoplasms prevention & control
- Abstract
Background: We conducted a study to determine whether dutasteride reduces the risk of incident prostate cancer, as detected on biopsy, among men who are at increased risk for the disease., Methods: In this 4-year, multicenter, randomized, double-blind, placebo-controlled, parallel-group study, we compared dutasteride, at a dose of 0.5 mg daily, with placebo. Men were eligible for inclusion in the study if they were 50 to 75 years of age, had a prostate-specific antigen (PSA) level of 2.5 to 10.0 ng per milliliter, and had had one negative prostate biopsy (6 to 12 cores) within 6 months before enrollment. Subjects underwent a 10-core transrectal ultrasound-guided biopsy at 2 and 4 years., Results: Among 6729 men who underwent a biopsy or prostate surgery, cancer was detected in 659 of the 3305 men in the dutasteride group, as compared with 858 of the 3424 men in the placebo group, representing a relative risk reduction with dutasteride of 22.8% (95% confidence interval, 15.2 to 29.8) over the 4-year study period (P<0.001). Overall, in years 1 through 4, among the 6706 men who underwent a needle biopsy, there were 220 tumors with a Gleason score of 7 to 10 among 3299 men in the dutasteride group and 233 among 3407 men in the placebo group (P=0.81). During years 3 and 4, there were 12 tumors with a Gleason score of 8 to 10 in the dutasteride group, as compared with only 1 in the placebo group (P=0.003). Dutasteride therapy, as compared with placebo, resulted in a reduction in the rate of acute urinary retention (1.6% vs. 6.7%, a 77.3% relative reduction). The incidence of adverse events was similar to that in studies of dutasteride therapy for benign prostatic hyperplasia, except that in our study, as compared with previous studies, the relative incidence of the composite category of cardiac failure was higher in the dutasteride group than in the placebo group (0.7% [30 men] vs. 0.4% [16 men], P=0.03)., Conclusions: Over the course of the 4-year study period, dutasteride reduced the risk of incident prostate cancer detected on biopsy and improved the outcomes related to benign prostatic hyperplasia. (ClinicalTrials.gov number, NCT00056407.), (2010 Massachusetts Medical Society)
- Published
- 2010
- Full Text
- View/download PDF
48. Oxidative stress and DNA methylation in prostate cancer.
- Author
-
Donkena KV, Young CY, and Tindall DJ
- Abstract
The protective effects of fruits, vegetables, and other foods on prostate cancer may be due to their antioxidant properties. An imbalance in the oxidative stress/antioxidant status is observed in prostate cancer patients. Genome oxidative damage in prostate cancer patients is associated with higher lipid peroxidation and lower antioxidant levels. Oxygen radicals are associated with different steps of carcinogenesis, including structural DNA damage, epigenetic changes, and protein and lipid alterations. Epigenetics affects genetic regulation, cellular differentiation, embryology, aging, cancer, and other diseases. DNA methylation is perhaps the most extensively studied epigenetic modification, which plays an important role in the regulation of gene expression and chromatin architecture, in association with histone modification and other chromatin-associated proteins. This review will provide a broad overview of the interplay of oxidative stress and DNA methylation, DNA methylation changes in regulation of gene expression, lifestyle changes for prostate cancer prevention, DNA methylation as biomarkers for prostate cancer, methods for detection of methylation, and clinical application of DNA methylation inhibitors for epigenetic therapy.
- Published
- 2010
- Full Text
- View/download PDF
49. Androgen regulation of gene expression.
- Author
-
Lamont KR and Tindall DJ
- Subjects
- Androgens genetics, Cell Differentiation, Cell Proliferation, Gene Targeting, Humans, Lipids biosynthesis, Male, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Receptors, Androgen metabolism, Androgens metabolism, Gene Expression Regulation, Neoplastic, Prostatic Neoplasms metabolism
- Abstract
The biological action of androgenic male sex steroid hormones in prostate tissue is mediated by the androgen receptor, a nuclear transcription factor. The transcriptional program of androgenic signaling in the prostate consists of thousands of gene targets whose products play a role in almost all cellular functions, including cellular proliferation, survival, lipid metabolism, and differentiation. This review will provide a summary of the most recent data regarding androgen-regulated target genes and modulation of androgen receptor activity, especially with regard to androgen-dependent and castration-recurrent prostate cancer., (Copyright (c) 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
50. New developments in the medical management of prostate cancer.
- Author
-
Kohli M and Tindall DJ
- Subjects
- Androgen Antagonists therapeutic use, Androgen Receptor Antagonists, Antineoplastic Agents therapeutic use, Humans, Immunotherapy, Male, Prostate-Specific Antigen blood, Prostatic Neoplasms diagnosis, Prostatic Neoplasms drug therapy, Prostatic Neoplasms therapy
- Abstract
Prostate cancer is a substantial public health burden and a leading cause of cancer-related morbidity and mortality in the United States despite the observation that annual prostate cancer-specific mortality rates have been declining during the previous decade. Although the reasons for this positive development are unclear, a combination of factors may have contributed. This update will review ongoing developments and summarize therapeutic advances in prostate cancer treatment on the basis of the current understanding of prostate cancer biology. Literature for this review was selected in 2009 by searching PubMed for the following keywords: prostatic neoplasms, castration, androgen receptor, hormonal, and chemotherapy. Emphasis is placed on published clinical studies in advanced prostate cancer therapeutics in the past 5 to 10 years. Also included in the review are novel hormonal agents targeting the androgen receptor currently in development for the treatment of advanced prostate cancer.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.