1
|
Lombe CP, Meyer M, Pretorius A. Bioinformatics Prediction and Analysis of MicroRNAs and Their Targets as Biomarkers for Prostate Cancer: A Preliminary Study. Mol Biotechnol 2021; 64:401-412. [PMID: 34665432 DOI: 10.1007/s12033-021-00414-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/06/2021] [Indexed: 12/24/2022]
Abstract
Prostate cancer (PCa) is the second most common form of cancer in men around the world. Due to its heterogeneity, presentations range from aggressive lethal disease to indolent disease. There is a need to identify core biomarkers that are important for early detection and progression, allowing a more precise method for the treatment and management of Pca. We obtained metastatic prostate cancer associated microRNA array profiles from the GSE28029 dataset in the GEO database. MicroRNA target prediction was done using the databases, TargetScanHuman, miRDB and DIANA microT, six target genes (FOXC1, CDKN1A, BIRC2, CTNND1, ELK1 and LRP8) were found to be common among the three different databases. Differential expression of the target genes was performed via the GENT2 database in the GPL96 platform (HG-U133A). Results indicated all genes were downregulated. Gene Ontology (GO) was used to perform enrichment analysis. The GO enrichment analysis indicated that the downregulated genes were enriched in cellular response to gamma radiation, regulation of transcription and response to drugs as well as protein binding and receptor signaling protein activity. The study suggested that CDKN1A, FOXC1 and BIRC2 might be core genes for prostate cancer that play an important role in its diagnosis, development and progression.
Collapse
Affiliation(s)
- Chipampe Patricia Lombe
- Department of Biotechnology, University of the Western Cape, Cape Town, South Africa. .,Biology Department, Mukuba University, Garneton itimpi, Kitwe, Zambia.
| | - Mervin Meyer
- Department of Biotechnology, University of the Western Cape, Cape Town, South Africa
| | - Ashley Pretorius
- Department of Biotechnology, University of the Western Cape, Cape Town, South Africa.,Biotechnology Innovation Division, Aminotek, PTY, Cape Town, South Africa
| |
Collapse
|
2
|
Singhal SS, Horne D, Singhal J, Awasthi S, Salgia R. Activating p53 function by targeting RLIP. Biochim Biophys Acta Rev Cancer 2021; 1875:188512. [PMID: 33460725 DOI: 10.1016/j.bbcan.2021.188512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 01/08/2023]
Abstract
Aberrations in RLIP, p53, and PKCα represent essentially the entire spectrum of all human neoplasms. Elevated PKCα expression, failure of the cell cycle checkpoint (p53 dysfunction), and abnormal glutathione (GSH) metabolism are fundamental hallmarks of carcinogenesis and drug/radiation resistance. However, a lack of investigations into the interactions between these important regulatory nodes has fundamentally limited our understanding of carcinogenesis and the development of effective interventions for cancer prevention and therapy. Loss of p53, perhaps the most powerful tumor suppressor gene, predisposes rodents to spontaneous cancer and humans to familial, as well as acquired, cancers. Until recently, no genetic manipulation of any oncogene had been reported to abrogate spontaneous carcinogenesis in p53-/- rodent models. However, the overexpression of RLIP, a GSH-electrophile conjugate (GS-E) transporter, has been found to enhance cancer cell proliferation and confer drug/radiation resistance, whereas its depletion causes tumor regression, suggesting its importance in cancer and drug/radiation resistance. Indeed, RLIP is an essential effector of p53 that is necessary for broad cancer-promoting epigenetic remodeling. Interestingly, through a haploinsufficiency mechanism, the partial depletion of RLIP in p53-/- mice provides complete protection from neoplasia. Furthermore, RLIP-/- mice exhibit altered p53 and PKCα function, marked deficiency in clathrin-dependent endocytosis (CDE), and almost total resistance to chemical carcinogenesis. Based on these findings, in this review, we present a novel and radical hypothesis that expands our understanding of the highly significant cross-talk between p53, PKCα, and GSH signaling by RLIP in multiple tumor models.
Collapse
Affiliation(s)
- Sharad S Singhal
- Department of Medical Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA.
| | - David Horne
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Jyotsana Singhal
- Department of Medical Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sanjay Awasthi
- Department of Internal Medicine, Division of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Ravi Salgia
- Department of Medical Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
3
|
Fan H, Hou J, Liu S, Xiao Z, Cui J. Long Non-Coding RNA DARS-AS1 Contributes to Prostate Cancer Progression Through Regulating the MicroRNA-628-5p/MTDH Axis. Cancer Manag Res 2020; 12:8363-8377. [PMID: 32982430 PMCID: PMC7490096 DOI: 10.2147/cmar.s271021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/19/2020] [Indexed: 12/18/2022] Open
Abstract
Purpose DARS antisense RNA 1 (DARS-AS1) is a long non-coding RNA that has been validated as a critical regulator in several human cancer types. Our study aimed to determine the expression profile of DARS-AS1 in prostate cancer (PCa) tissues and cell lines. Functional experiments were conducted to explore the detailed roles of DARS-AS1 in regulating PCa carcinogenesis. Furthermore, the detailed mechanisms by which DARS-AS1 regulates the oncogenicity of PCa cells were uncovered. Methods Reverse transcription quantitative polymerase chain reaction was performed to analyze DARS-AS1 expression in PCa tissues and cell lines. Cell Counting Kit-8 assays, flow cytometry analyses, Transwell assays, and tumor xenograft experiments were conducted to determine the regulatory effects of DARS-AS1 knockdown on the malignant phenotype of PCa cells. Bioinformatics analysis was performed to identify putative microRNAs (miRNAs) targeting DARS-AS1, and the direct interaction between DARS-AS1 and miR-628-5p was verified using RNA immunoprecipitation and luciferase reporter assays. Results DARS-AS1 was highly expressed in PCa tissues and cell lines. In vitro functional experiments demonstrated that DARS-AS1 depletion suppressed PCa cell proliferation, promoted cell apoptosis, and restricted cell migration and invasion. In vivo studies revealed that the downregulation of DARS-AS1 inhibited PCa tumor growth in nude mice. Mechanistic investigation verified that DARS-AS1 functioned as an endogenous miR-628-5p sponge in PCa cells and consequently promoted the expression of metadherin (MTDH). Furthermore, the involvement of miR-628-5p/MTDH axis in DARS-AS1-mediated regulatory actions in PCa cells was verified using rescue experiments. Conclusion DARS-AS1 functioned as a competing endogenous RNA in PCa by adsorbing miR-628-5p and thereby increasing the expression of MTDH, resulting in enhanced PCa progression. The identification of a novel DARS-AS1/miR-628-5p/MTDH regulatory network in PCa cells may offer a new theoretical basis for the development of promising therapeutic targets.
Collapse
Affiliation(s)
- Haitao Fan
- Department of Urology, The Second Hospital of Jilin University, Changchun, Jilin 130041, People's Republic of China
| | - Junhui Hou
- Department of Oncology & Radiotherapy, Qingdao Central Medical Group, Qingdao, Shandong 266000, People's Republic of China
| | - Siqing Liu
- Department of Outpatient, Qingdao Special Service Sanatorium of PLA Navy, Qingdao, Shandong 266071, People's Republic of China
| | - Zuomin Xiao
- Department of Clinical Laboratory, Jinan Jigang Hospital, Jinan, Shandong 250101, People's Republic of China
| | - Jia Cui
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, People's Republic of China
| |
Collapse
|
4
|
Shukla S, Srivastava JK, Shankar E, Kanwal R, Nawab A, Sharma H, Bhaskaran N, Ponsky LE, Fu P, MacLennan GT, Gupta S. Oxidative Stress and Antioxidant Status in High-Risk Prostate Cancer Subjects. Diagnostics (Basel) 2020; 10:E126. [PMID: 32120827 PMCID: PMC7151307 DOI: 10.3390/diagnostics10030126] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 02/21/2020] [Accepted: 02/24/2020] [Indexed: 12/24/2022] Open
Abstract
The oxidant/antioxidant balance has been implicated in the pathophysiology of prostate cancer. We investigated oxidative damage and antioxidant status in high-risk prostate cancer subjects. Reduced glutathione (GSH) levels were measured in erythrocytes, 8-hydroxydeoxyguanosine (8-OHdG) in leukocytes and plasma levels of catalase (CAT), glutathione peroxidase (GSH-Px), glutathione reductase (GSH-R), glutathione S-transferase (GST), superoxide dismutase (SOD), and lipid peroxide products were measured in high-risk and age-matched healthy subjects. Serum PSA levels were significantly higher (p < 0.0001) in high-risk subjects, whereas GST (p < 0.0001) and GSH (p < 0.002) were higher in healthy controls. Levels of 8-OHdG, an oxidized nucleoside of DNA, were significantly increased (p < 0.0001) in high-risk subjects. No marked difference in the levels of CAT (p = 0.237), GSH-Px (p = 0.74), GSH-R (p = 0.344), SOD (p = 0.109), and lipid peroxide products (p = 0129) were observed between two groups. Pearson's correlation between GST and PSA (r = -0.69 (p < 0.0001)), GST and 8-OHdG (r = -0.62 (p < 0.0004)), GSH and 8-OHdG (r= -0.39 (p = 0.038)), and CAT and GSH-Px (r= -0.33 (p = 0.04)) were found to be negatively correlated, whereas 8-OHdG and PSA were positively associated (r= 0.57 (p < 0.002). These results indicate a significant role of oxidative damage in prostate carcinogenesis, particularly during the early stages of development. In conclusion, our data support the importance of antioxidant defense as a valuable diagnostic and/or prognostic marker in prostate cancer.
Collapse
Affiliation(s)
- Sanjeev Shukla
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA; (S.S.); (J.K.S.); (E.S.); (R.K.); (A.N.); (H.S.); (N.B.); (L.E.P.)
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA;
| | - Janmejai K. Srivastava
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA; (S.S.); (J.K.S.); (E.S.); (R.K.); (A.N.); (H.S.); (N.B.); (L.E.P.)
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA;
| | - Eswar Shankar
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA; (S.S.); (J.K.S.); (E.S.); (R.K.); (A.N.); (H.S.); (N.B.); (L.E.P.)
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA;
| | - Rajnee Kanwal
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA; (S.S.); (J.K.S.); (E.S.); (R.K.); (A.N.); (H.S.); (N.B.); (L.E.P.)
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA;
| | - Akbar Nawab
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA; (S.S.); (J.K.S.); (E.S.); (R.K.); (A.N.); (H.S.); (N.B.); (L.E.P.)
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA;
- Present Address: Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32611 USA
| | - Haripaul Sharma
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA; (S.S.); (J.K.S.); (E.S.); (R.K.); (A.N.); (H.S.); (N.B.); (L.E.P.)
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA;
| | - Natarajan Bhaskaran
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA; (S.S.); (J.K.S.); (E.S.); (R.K.); (A.N.); (H.S.); (N.B.); (L.E.P.)
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA;
| | - Lee E. Ponsky
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA; (S.S.); (J.K.S.); (E.S.); (R.K.); (A.N.); (H.S.); (N.B.); (L.E.P.)
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA;
| | - Pingfu Fu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Gregory T. MacLennan
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA;
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Sanjay Gupta
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA; (S.S.); (J.K.S.); (E.S.); (R.K.); (A.N.); (H.S.); (N.B.); (L.E.P.)
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA;
- Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106, USA
- Division of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
- Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
5
|
ZEB1 activated-VPS9D1-AS1 promotes the tumorigenesis and progression of prostate cancer by sponging miR-4739 to upregulate MEF2D. Biomed Pharmacother 2019; 122:109557. [PMID: 31918265 DOI: 10.1016/j.biopha.2019.109557] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 10/15/2019] [Accepted: 10/17/2019] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (PCa) is a destructive malignancy with a bad prognosis. LncRNA VPS9D1-AS1 has recently been delineated as an oncogene in some kinds of tumor, whereas, the function of VPS9D1-AS1 in PCa remains to be clarified. In this study, we researched its underlying role in PCa. The expression of VPS9D1-AS1 was conspicuously upregulated in PCa tissues and cells. And absence of VPS9D1-AS1 inhibited cell proliferation, migration and invasion, and promoted cell apoptosis in PCa. In addition, VPS9D1-AS1 overexpression led to opposite results. Furthermore, VPS9D1-AS1/MEF2D could sponge with miR-4739. VPS9D1-AS1/MEF2D and miR-4739 were inversely correlated in tumor cells. And the expression of miR-4739 is markedly downregulated in PCa, meanwhile, that of MEF2D exhibited the opposite tendency. However, MEF2D was positively regulated by VPS9D1-AS1. Moreover, MEF2D upregulation offset the suppressive effects of VPS9D1-AS1 deficiency on cell proliferation, migration and invasion in PCa. Additionally, ZEB1 contained the binding sites of VPS9D1-AS1 promoter, and there existed positive relation between them. Taken together, above results illustrated that ZEB1 activated-VPS9D1-AS1 promotes the tumorigenesis and progression of PCa by sponging miR-4739 to upregulate MEF2D, which offering a new useful reference for studying the development process of PCa.
Collapse
|
6
|
Li B, Guo Z, Liang Q, Zhou H, Luo Y, He S, Lin Z. lncRNA DGCR5 Up-Regulates TGF-β1, Increases Cancer Cell Stemness and Predicts Survival of Prostate Cancer Patients. Cancer Manag Res 2019; 11:10657-10663. [PMID: 31920375 PMCID: PMC6939399 DOI: 10.2147/cmar.s231112] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 11/14/2019] [Indexed: 11/24/2022] Open
Abstract
Background Long non-coding RNA (lncRNA) DiGeorge syndrome critical region gene 5 (DGCR5) plays different roles in different types of human cancer, but its role in prostate cancer (PC) has not been reported. Methods DGCR5 and TGF-β1 expression in paired tumor and adjacent healthy tissues from 64 PC patients was analyzed by performing RT-qPCR. A 5-year follow-up study was performed to analyze the prognostic value of DGCR5 for PC. The interaction between DGCR5 and TGF-β1 was analyzed by overexpression experiments. Cell stemness was analyzed by cell stemness assay. Results In our study, we found that DGCR5 was down-regulated in tumor tissues than in adjacent healthy tissues of PC patients, but TGF-β1 was up-regulated in the tumor tissues. DGCR5 expression was not affected by clinical stages, but low DGCR5 level in the tumor was correlated with poor survival. DGCR5 and TGF-β1 were inversely correlated in tumor tissues but not in adjacent healthy tissues. DGCR5 over-expression resulted in down-regulation of TGF-β1, while TGF-β1 treatment did not significantly affect DGCR5 expression. DGCR5 over-expression led to decreased stemness of PC cells, but TGF-β1 treatment played a reverse role and attenuated the effects of DGCR5 over-expression. DGCR5 may decrease the stemness of PC cells by down-regulating TGF-β1.
Collapse
Affiliation(s)
- Bin Li
- Department of Urology, The First Peoples' Hospital of Foshan, Foshan City, Guangdong Province 528000, People's Republic of China
| | - Zhirui Guo
- Department of Radiology, The First Peoples' Hospital of Foshan, Foshan City, Guangdong Province 528000, People's Republic of China
| | - Quan Liang
- Department of Urology, The First Peoples' Hospital of Foshan, Foshan City, Guangdong Province 528000, People's Republic of China
| | - Huiling Zhou
- Department of Infectious Disease, The First Peoples' Hospital of Foshan, Foshan City, Guangdong Province 528000, People's Republic of China
| | - Yanping Luo
- Department of Anesthesiology Surgery, The First Peoples' Hospital of Foshan, Foshan City, Guangdong Province 528000, People's Republic of China
| | - Shuyun He
- Department of Radiology, The First Peoples' Hospital of Foshan, Foshan City, Guangdong Province 528000, People's Republic of China
| | - Zhe Lin
- Department of Urology, The First Peoples' Hospital of Foshan, Foshan City, Guangdong Province 528000, People's Republic of China
| |
Collapse
|
7
|
Wu Z, Huang W, Chen Y, Chen B, Liu R, Bai P, Xing J. LINC01638 lncRNA promotes the proliferation, migration and invasion of prostate carcinoma cells by interacting with Notch1. Cancer Biomark 2019; 25:161-168. [PMID: 31104008 DOI: 10.3233/cbm-182137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
LINC01638 lncRNA is known as an oncogenic lncRNA in triple negative breast cancer. However, the role of LINC01638 lncRNA in other diseases is unknown. In the present study we observed that plasma levels of LINC01638 lncRNA and Notch1 were upregulated in prostate carcinoma patients comparing with healthy controls. LINC01638 lncRNA and Notch1 were positively correlated in prostate carcinoma patients but not in healthy controls. Upregulation of LINC01638 lncRNA distinguished prostate carcinoma patients from healthy controls. LINC01638 lncRNA overexpression in prostate carcinoma cells led to upregulated Notch1 expression. Notch1 overexpression also led to increased expression level of LINC01638 lncRNA. Both LINC01638 lncRNA and Notch1 overexpression promoted the proliferation, migration and invasion of prostate carcinoma cells. We concluded that LINC01638 lncRNA might promote the proliferation, migration and invasion of prostate carcinoma cells by interacting with Notch1.
Collapse
|
8
|
Li G, Zhang Y, Mao J, Hu P, Chen Q, Ding W, Pu R. LncRNA TUC338 is overexpressed in prostate carcinoma and downregulates miR-466. Gene 2019; 707:224-230. [PMID: 31085276 DOI: 10.1016/j.gene.2019.05.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 12/20/2022]
Abstract
LncRNA TUC338 has recently been characterized as an oncogene in several types of cancer. Our study aimed to characterize the functionality of TUC338 in prostate carcinoma. It was observed that TUC338 was upregulated in tumor tissues comparing to adjacent healthy tissues of prostate carcinoma patients. Plasma levels of TUC338 were also higher in prostate carcinoma patients than in healthy controls. A 5-year follow-up study showed that high plasma level of TUC338 was correlated with poor survival. miR-466 was downregulated in tumor tissues compared with adjacent healthy tissues of prostate carcinoma patients. TUC338 and miR-466 were inversely correlated in tumor tissues. miR-466 overexpression failed to affect TUC338 expression, while TUC338 overexpression led to downregulated miR-466 expression. TUC338 overexpression failed to significantly affect cancer cell proliferation, but promoted cancer cell migration and invasion. MiR-466 overexpression resulted in reduced rates of cancer cell migration and invasion, and also attenuated the effect of TUC338 overexpression. Therefore, TUC338 may serve as an oncogenic lncRNA in prostate carcinoma by downregulating miR-466.
Collapse
Affiliation(s)
- Gang Li
- Department of Urology, Mianzhu City People's Hospital, Mianzhu City, Sichuan Province 618200, PR China
| | - Yang Zhang
- Department of Urology, Mianzhu City People's Hospital, Mianzhu City, Sichuan Province 618200, PR China
| | - Jing Mao
- Department of Urology, Mianzhu City People's Hospital, Mianzhu City, Sichuan Province 618200, PR China
| | - Peng Hu
- Department of Urology, Mianzhu City People's Hospital, Mianzhu City, Sichuan Province 618200, PR China
| | - Qiang Chen
- Department of Urology, Mianzhu City People's Hospital, Mianzhu City, Sichuan Province 618200, PR China
| | - Wei Ding
- Department of Urology, Mianzhu City People's Hospital, Mianzhu City, Sichuan Province 618200, PR China
| | - Rong Pu
- Department of Laboratory,the third people's Hospital of Dongguan City, Guangdong Dongguan 523326,PR China.
| |
Collapse
|
9
|
The multifaceted role of glutathione S-transferases in cancer. Cancer Lett 2018; 433:33-42. [PMID: 29959055 DOI: 10.1016/j.canlet.2018.06.028] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 02/07/2023]
Abstract
Glutathione S-transferases (GSTs) are phase II detoxifying enzymes involved in the maintenance of cell integrity, oxidative stress and protection against DNA damage by catalyzing the conjugation of glutathione to a wide variety of electrophilic substrates. Though enzymes of the glutathione synthesis and salvage pathways have been well characterized in the past, there is still a lack of comprehensive understanding of their independent and coordinate regulatory mechanisms in carcinogenesis. The present review discusses implication of GST in cancer development and progression, gene polymorphism, drug resistance, signaling and epigenetic regulation involving their role in cancer. It is anticipated that GST especially the GSTP1 class can be developed as a biomarker either used alone or in combination with other biomarkers for early cancer detection and/or diagnosis as well as for future targeted preventive and therapeutic interventions with dietary agents.
Collapse
|
10
|
Nodouzi V, Nowroozi M, Hashemi M, Javadi G, Mahdian R. Concurrent Down-Regulation of PTEN and NKX3.1 Expression in Iranian Patients with Prostate Cancer. Int Braz J Urol 2016; 41:898-905. [PMID: 26689514 PMCID: PMC4756965 DOI: 10.1590/s1677-5538.ibju.2014.0036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 05/22/2014] [Indexed: 11/22/2022] Open
Abstract
NKX3.1 and PTEN genes are involved in the development and progression of prostate cancer (PCa). Here, in line with other studies that correlated the expression of these two genes, we aimed at evaluating the expression pattern of these genes in clinical PCa samples. Collectively, 81 tissue samples including 45 human PCa and 36 benign prostatic hyperplasia (BPH) specimens were included in the study. The tissue samples were subjected to RNA extraction and subsequently to cDNA synthesis according to the kit manufacturer's protocol. Quantitative Real-Time PCR assay was performed for each sample in triplicate reactions. REST and SPSS software were used to statistically analyze PTEN and NKX3.1 gene expression data. Expression level of both NKX3.1 and PTEN genes was down-regulated in PCa samples compared to BPH samples. The relative expression ratio of PTEN and NKX3.1 was decreased to 0.155 and 0.003, respectively (P=0.000). The results of Chi-Square analysis revealed a significant correlation between the expression of these genes in both BPH and cancer groups (P=0.004 and 0.001, respectively). According to previous studies and our data, we concluded that the association between the down-regulation of PTEN and NKX3.1 genes contributed to the prostate tumorigenesis. This might highlight the interaction between the proteins encoded by these genes. Furthermore, this finding might be exploited for the development of innovative diagnostic and therapeutic approaches in PCa.
Collapse
Affiliation(s)
- Vahideh Nodouzi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Mehrdad Hashemi
- Department of Genetics Tehran Medical Branch, Islamic Azad University, Tehran, Iran
| | - Gholareza Javadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Reza Mahdian
- Department of Molecular Medicine, Pasteur Institute of Iran, Biotechnology Research Center, Tehran, Iran
| |
Collapse
|
11
|
Mian OY, Khattab MH, Hedayati M, Coulter J, Abubaker-Sharif B, Schwaninger JM, Veeraswamy RK, Brooks JD, Hopkins L, Shinohara DB, Cornblatt B, Nelson WG, Yegnasubramanian S, DeWeese TL. GSTP1 Loss results in accumulation of oxidative DNA base damage and promotes prostate cancer cell survival following exposure to protracted oxidative stress. Prostate 2016; 76:199-206. [PMID: 26447830 PMCID: PMC4734373 DOI: 10.1002/pros.23111] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 09/28/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Epigenetic silencing of glutathione S-transferase π (GSTP1) is a hallmark of transformation from normal prostatic epithelium to adenocarcinoma of the prostate. The functional significance of this loss is incompletely understood. The present study explores the effects of restored GSTP1 expression on glutathione levels, accumulation of oxidative DNA damage, and prostate cancer cell survival following oxidative stress induced by protracted, low dose rate ionizing radiation (LDR). METHODS GSTP1 protein expression was stably restored in LNCaP prostate cancer cells. The effect of GSTP1 restoration on protracted LDR-induced oxidative DNA damage was measured by GC-MS quantitation of modified bases. Reduced and oxidized glutathione levels were measured in control and GSTP1 expressing populations. Clonogenic survival studies of GSTP1- transfected LNCaP cells after exposure to protracted LDR were performed. Global gene expression profiling and pathway analysis were performed. RESULTS GSTP1 expressing cells accumulated less oxidized DNA base damage and exhibited decreased survival compared to control LNCaP-Neo cells following oxidative injury induced by protracted LDR. Restoration of GSTP1 expression resulted in changes in modified glutathione levels that correlated with GSTP1 protein levels in response to protracted LDR-induced oxidative stress. Survival differences were not attributable to depletion of cellular glutathione stores. Gene expression profiling and pathway analysis following GSTP1 restoration suggests this protein plays a key role in regulating prostate cancer cell survival. CONCLUSIONS The ubiquitous epigenetic silencing of GSTP1 in prostate cancer results in enhanced survival and accumulation of potentially promutagenic DNA adducts following exposure of cells to protracted oxidative injury suggesting a protective, anti-neoplastic function of GSTP1. The present work provides mechanistic backing to the tumor suppressor function of GSTP1 and its role in prostate carcinogenesis.
Collapse
Affiliation(s)
- Omar Y. Mian
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland
| | - Mohamed H. Khattab
- School of Medicine, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland
| | - Mohammad Hedayati
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland
| | - Jonathan Coulter
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland
| | - Budri Abubaker-Sharif
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland
- School of Medicine, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland
| | | | | | - James D. Brooks
- Department of Urology, Stanford University, Palo Alto, California
| | - Lisa Hopkins
- St. Peters University Hospital Breast Center in New Brunswick, New Jersey
| | - Debika Biswal Shinohara
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland
| | | | - William G. Nelson
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland
| | - Srinivasan Yegnasubramanian
- School of Medicine, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland
| | - Theodore L. DeWeese
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland
- School of Medicine, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland
- Correspondence to: Theodore L. DeWeese, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland 21287-5678.
| |
Collapse
|
12
|
WANG JIACHEN, WANG ZHAO, FAN YUXIA, SI YAQING, WANG JIAXIANG. DNA methyltransferase 3b silencing affects locus-specific DNA methylation and inhibits proliferation, migration and invasion in human hepatocellular carcinoma SMMC-7721 and BEL-7402 cells. Oncol Lett 2015; 9:2499-2506. [PMID: 26137097 PMCID: PMC4473378 DOI: 10.3892/ol.2015.3077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 03/06/2015] [Indexed: 12/22/2022] Open
Abstract
DNA methylation is an important regulator of gene transcription, and its role in carcinogenesis has been a topic of considerable interest in previous years. The present study examined the influence of DNA methyltransferase 3b (DNMT3b) on cell proliferation, migration and invasion, and the methylation status of identified tumor suppressor genes in hepatoma SMMC-7721 and BEL-7402 cells. DNMT3b was silenced by small interfering RNA (siRNA) in human hepatocellular carcinoma cell lines. Transfection efficiency was verified using a fluorescent imaging system, reverse transcription polymerase chain reaction (RT-PCR) and western blotting. A cell proliferation assay was performed to evaluate cell viability. Cell cycle distribution and apoptosis were analyzed by flow cytometry. The migratory and invasive ability of cells was measured using a Transwell assay. Methylation-specific PCR (MSP) was performed to assess methylation in the promoter region of genes. The present data revealed that DNMT3b siRNA successfully inhibited expression of the DNMT3b gene in these two liver cancer cell lines and therefore inhibited the proliferation of the transfected cells, stimulated apoptosis in the cells, led to an accumulation of cells in the G2/M phase and decreased cell migration and invasion. It was also found that silencing DNMT3b expression results in hypomethylation of specific sets of gene promoters and increases the expression of distinct set of genes in HCC cell lines. The present study is therefore useful for assessing the specificity of emerging action based on the altered expression of associated regulatory genes, particularly in methylation-silenced genes.
Collapse
Affiliation(s)
- JIA-CHEN WANG
- Department of Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - ZHAO WANG
- Department of Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - YU-XIA FAN
- Department of Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - YA-QING SI
- Department of Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - JIA-XIANG WANG
- Department of Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
13
|
Jurado-Penagos A, Garcia-Perdomo HA. Efectividad de los micronutrientes en la prevención del cáncer de próstata. REVISTA DE LA FACULTAD DE MEDICINA 2015. [DOI: 10.15446/revfacmed.v62n3sup.44209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
14
|
Gecit I, Meral I, Aslan M, Kocyigit A, Celik H, Taskın A, Kaba M, Pirincci N, Gunes M, Taken K, Demir H, Uyuklu M, Ceylan K. Peripheral mononuclear leukocyte DNA damage, plasma prolidase activity, and oxidative status in patients with benign prostatic hyperplasia. Redox Rep 2015; 20:163-9. [PMID: 25551736 DOI: 10.1179/1351000214y.0000000121] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
OBJECTIVES Prolidase plays a major role in collagen turnover, matrix remodeling, and cell growth. Benign prostatic hyperplasia (BPH) may be associated with an increased extracellular matrix deposition. Therefore, the present study was designed to investigate the plasma prolidase activity, oxidative status, and peripheral mononuclear leukocyte DNA damage in patients with BPH. PATIENTS AND METHODS Twenty-six male patients with BPH and 24 healthy male subjects were included in this study. Blood samples were collected from antecubital vein after an overnight fasting period, and the plasma was separated. Plasma prolidase activity, total antioxidant capacity (TAC), total oxidant status (TOS), and oxidative stress index (OSI) were determined. The peripheral lymphocyte oxidative DNA damage was determined using an alkaline single cell gel electrophoresis assay (comet assay). RESULTS The plasma prolidase activity, TOS levels, OSI values, and peripheral mononuclear leukocyte DNA damage were significantly higher (P < 0.001), while the TAC levels were significantly lower (P < 0.001) in patients with BPH than controls. In BPH patients, the prolidase activity was significantly associated with TAC levels (r = -0.366, P < 0.05), TOS levels (r = 0.573, P < 0.001), and OSI (r = 0.618, P < 0.001) and peripheral mononuclear leukocyte DNA damage (r = 0.461, P < 0.001). CONCLUSIONS Our results showed that BPH might be associated with an increased oxidative stress, and also an increased plasma prolidase activity. Increased prolidase activity might play an important role in the etiopathogenesis and/or progression of BPH.
Collapse
|
15
|
Hahn AM, Myers JD, McFarland EK, Lee S, Jerde TJ. Interleukin-driven insulin-like growth factor promotes prostatic inflammatory hyperplasia. J Pharmacol Exp Ther 2014; 351:605-15. [PMID: 25292180 PMCID: PMC4244580 DOI: 10.1124/jpet.114.218693] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Accepted: 10/03/2014] [Indexed: 01/30/2023] Open
Abstract
Prostatic inflammation is of considerable importance to urologic research because of its association with benign prostatic hyperplasia and prostate cancer. However, the mechanisms by which inflammation leads to proliferation and growth remain obscure. Here, we show that insulin-like growth factors (IGFs), previously known as critical developmental growth factors during prostate organogenesis, are induced by inflammation as part of the proliferative recovery to inflammation. Using genetic models and in vivo IGF receptor blockade, we demonstrate that the hyperplastic response to inflammation depends on interleukin-1-driven IGF signaling. We show that human prostatic hyperplasia is associated with IGF pathway activation specifically localized to foci of inflammation. This demonstrates that mechanisms of inflammation-induced epithelial proliferation and hyperplasia involve the induction of developmental growth factors, further establishing a link between inflammatory and developmental signals and providing a mechanistic basis for the management of proliferative diseases by IGF pathway modulation.
Collapse
Affiliation(s)
- Alana M Hahn
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana (A.M.H., J.D.M., T.J.J.); Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin (E.K.M., S.L.); and Melvin and Bren Simon Cancer Center, Indiana Basic Urological Research Working Group, Indiana University, Indianapolis, Indiana (T.J.J.)
| | - Jason D Myers
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana (A.M.H., J.D.M., T.J.J.); Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin (E.K.M., S.L.); and Melvin and Bren Simon Cancer Center, Indiana Basic Urological Research Working Group, Indiana University, Indianapolis, Indiana (T.J.J.)
| | - Eliza K McFarland
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana (A.M.H., J.D.M., T.J.J.); Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin (E.K.M., S.L.); and Melvin and Bren Simon Cancer Center, Indiana Basic Urological Research Working Group, Indiana University, Indianapolis, Indiana (T.J.J.)
| | - Sanghee Lee
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana (A.M.H., J.D.M., T.J.J.); Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin (E.K.M., S.L.); and Melvin and Bren Simon Cancer Center, Indiana Basic Urological Research Working Group, Indiana University, Indianapolis, Indiana (T.J.J.)
| | - Travis J Jerde
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana (A.M.H., J.D.M., T.J.J.); Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin (E.K.M., S.L.); and Melvin and Bren Simon Cancer Center, Indiana Basic Urological Research Working Group, Indiana University, Indianapolis, Indiana (T.J.J.)
| |
Collapse
|
16
|
McPartlin DA, O'Kennedy RJ. Point-of-care diagnostics, a major opportunity for change in traditional diagnostic approaches: potential and limitations. Expert Rev Mol Diagn 2014; 14:979-98. [PMID: 25300742 DOI: 10.1586/14737159.2014.960516] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
'Point-of-care' (POC) diagnostics are a powerful emerging healthcare approach. They can rapidly provide statistically significant results, are simple to use, do not require specialized equipment and are cost-effective. For these reasons, they have the potential to play a major role in revolutionizing the diagnosis, initiation and monitoring of treatment of major global diseases. This review focuses on antibody-based POC devices that target four major global diseases: cardiovascular diseases, prostate cancer, HIV infection and tuberculosis. The key statistics and pathology of each disease is described in detail, followed by an in-depth discussion on emerging POC devices that target each disease, highlighting their potential and limitations.
Collapse
Affiliation(s)
- Daniel A McPartlin
- School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Co. Dublin, Ireland
| | | |
Collapse
|
17
|
Naiki T, Asamoto M, Toyoda-Hokaiwado N, Naiki-Ito A, Tozawa K, Kohri K, Takahashi S, Shirai T. Organ specific Gst-pi expression of the metastatic androgen independent prostate cancer cells in nude mice. Prostate 2012; 72:533-41. [PMID: 21748757 DOI: 10.1002/pros.21455] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Accepted: 06/15/2011] [Indexed: 11/08/2022]
Abstract
BACKGROUND Elucidating the mechanisms of metastasis in prostate cancer, particularly to the bone, is a major issue for treatment of this malignancy. We previously reported that an androgen-independent variant had higher expression of glutathione S-transferase pi (Gst-pi) compared with a parent androgen-dependent transplantable rat prostate carcinoma which was established from the transgenic rat for adenocarcinoma of the prostate (TRAP). METHODS A new cell line, PCai1, was established from the androgen-independent tumor and investigated its metastatic potential in nude mice. The tumorigenesis of PCai1 cells in vivo was studied by subcutaneous transplantations into nude mice. The growth in the microenvironment of the prostate was studied by orthotopic transplantation of PCai1 cells into nude mice. The metastatic potential of PCai1 cells was studied by tail vein injections. Effects of Gst-pi knocked down were analysis in PCai1 cells. RESULTS PCai1 frequently formed metastatic lesions in the lung and lymph nodes after orthotopic implantation in the prostate. Intravenous injections of PCai1, metastasis to lung and bone were obvious. PCai1 had strong expression for Gst-pi, therefore we tried knocked down Gst-pi. Gst-pi-siRNA in vitro significantly suppressed cell proliferation rate. In addition, high levels of intracellular reactive oxygen species (ROS) were recognized in the Gst-pi knockout. CONCLUSIONS Gst-pi expression of the prostate cancers are dependent on metastatic site, and that Gst-pi has an important role in adapting prostate cancer for growth and metastasis involving an alteration of ROS signals.
Collapse
Affiliation(s)
- Taku Naiki
- Department of Experimental Pathology and Tumor Biology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Cancer patients' outcome and survival depends on the early diagnosis of malignant lesions. Several investigation methods used for the prevention and early detection strategies have specific limitations. More recently, epigenetic changes have been considered one of the most promising tools for the early diagnosis of cancer. Some of these epigenetic alterations including promoter hypermethylation of genes like P16INK4a, BRCA1, BRCA2, ERα and RARβ2, APC, and RASSF1A have been associated with early stages of mammary gland tumorigenesis and have been suggested to be included in the models that evaluate individual breast cancer risk. In lung cancer, P16INK4a and MGMT gene hypermethylation was observed in sputum years before clinical manifestation of the squamous cell carcinoma among smokers. Loss of GSTP1 function by DNA hypermethylation together with changes in the methylation levels of repetitive elements like LINE-1 and Sat2 was reported in prostatic preneoplastic lesions. Also, DNA hypermethylation for hMLH1 and MGMT DNA repair genes was reported in precursor lesions to colorectal cancer. These epigenetic alterations may be influenced by factors such as xenoestrogens, folate, and multivitamins. Detection of these changes may help determining cancer susceptibility and early diagnosis.
Collapse
|
19
|
Barry KH, Koutros S, Berndt SI, Andreotti G, Hoppin JA, Sandler DP, Burdette LA, Yeager M, Freeman LEB, Lubin JH, Ma X, Zheng T, Alavanja MCR. Genetic variation in base excision repair pathway genes, pesticide exposure, and prostate cancer risk. ENVIRONMENTAL HEALTH PERSPECTIVES 2011; 119:1726-1732. [PMID: 21810555 PMCID: PMC3261977 DOI: 10.1289/ehp.1103454] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 08/02/2011] [Indexed: 05/30/2023]
Abstract
BACKGROUND Previous research indicates increased prostate cancer risk for pesticide applicators and pesticide manufacturing workers. Although underlying mechanisms are unknown, evidence suggests a role of oxidative DNA damage. OBJECTIVES Because base excision repair (BER) is the predominant pathway involved in repairing oxidative damage, we evaluated interactions between 39 pesticides and 394 tag single-nucleotide polymorphisms (SNPs) for 31 BER genes among 776 prostate cancer cases and 1,444 male controls in a nested case-control study of white Agricultural Health Study (AHS) pesticide applicators. METHODS We used likelihood ratio tests from logistic regression models to determine p-values for interactions between three-level pesticide exposure variables (none/low/high) and SNPs (assuming a dominant model), and the false discovery rate (FDR) multiple comparison adjustment approach. RESULTS The interaction between fonofos and rs1983132 in NEIL3 [nei endonuclease VIII-like 3 (Escherichia coli)], which encodes a glycosylase that can initiate BER, was the most significant overall [interaction p-value (pinteract) = 9.3 × 10-6; FDR-adjusted p-value = 0.01]. Fonofos exposure was associated with a monotonic increase in prostate cancer risk among men with CT/TT genotypes for rs1983132 [odds ratios (95% confidence intervals) for low and high use compared with no use were 1.65 (0.91, 3.01) and 3.25 (1.78, 5.92), respectively], whereas fonofos was not associated with prostate cancer risk among men with the CC genotype. Carbofuran and S-ethyl dipropylthiocarbamate (EPTC) interacted similarly with rs1983132; however, these interactions did not meet an FDR < 0.2. CONCLUSIONS Our significant finding regarding fonofos is consistent with previous AHS findings of increased prostate cancer risk with fonofos exposure among those with a family history of prostate cancer. Although requiring replication, our findings suggest a role of BER genetic variation in pesticide-associated prostate cancer risk.
Collapse
Affiliation(s)
- Kathryn Hughes Barry
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Rockville, Maryland 20892-7240, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Zecha H, Schmid HP, Tschopp A, Sulser T, Engeler DS. High incidence of independent second malignancies after non-muscle-invasive bladder cancer. ACTA ACUST UNITED AC 2011; 45:245-50. [DOI: 10.3109/00365599.2011.562234] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Henrik Zecha
- Department of Urology,
St. Gallen Cantonal Hospital, St. Gallen, Switzerland
| | - Hans-Peter Schmid
- Department of Urology,
St. Gallen Cantonal Hospital, St. Gallen, Switzerland
| | - Alois Tschopp
- Biostatistics Unit, Institute for Social and Preventive Medicine,
University of Zurich, Zurich, Switzerland
| | - Tullio Sulser
- Department of Urology,
University Hospital Zurich, Zurich, Switzerland
| | - Daniel S. Engeler
- Department of Urology,
University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
21
|
Bushman W. Etiology, epidemiology, and natural history of benign prostatic hyperplasia. Urol Clin North Am 2010; 36:403-15, v. [PMID: 19942041 DOI: 10.1016/j.ucl.2009.07.003] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Historically, benign prostatic hyperplasia (BPH) has been a major focus of urologic practice and surgery. But a simplistic causal relationship among prostatic enlargement, progressive obstruction, lower urinary tract symptoms, retention, and complications of retention has been challenged by recognition of the incomplete overlap of prostatic enlargement with symptoms and obstruction. The result has been a greater focus on symptoms than prostatic enlargement and a shift from surgery to medical treatment. Therefore, the question can be asked whether BPH per se, the glandular enlargement as it contributes to bladder dysfunction, or hyperplastic enlargement as a biomarker for generalized lower urinary tract dysfunction are concerns. This article addresses these issues.
Collapse
Affiliation(s)
- Wade Bushman
- Department of Urology, University of Wisconsin Medical School, K6-562 Clinical Science Center, 600 Highland Avenue, Madison, WI 53792, USA.
| |
Collapse
|
22
|
Singh RK, Sudhakar A, Lokeshwar BL. Role of Chemokines and Chemokine Receptors in Prostate Cancer Development and Progression. ACTA ACUST UNITED AC 2010; 2:89-94. [PMID: 20808724 DOI: 10.4172/1948-5956.1000030] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Prostate cancer (PC) is the second leading cause of cancer deaths in men in America and Western Europe. Epidemiological studies suggest that prostate cancer incidences increased in last few years in Asian. The causes or consequences of increasing trend of prostate cancer incidence are not completely known. Emerging evidences suggest that among the many risk factors, inflammation is the major risk factor for developing prostate cancer and its progression to metastasis. It is proposed that exposure to environmental factors such as infectious agents, dietary agents and saturated lipids leads to injury of the prostate due to chronic inflammation and regenerative risk factor lesions referred to as proliferative inflammatory atrophy (PIA). These phenomena predominantly control by a number of proinflammatory macro molecules such as chemokines, and their receptors. Some recent studies suggest that many of these pro-inflammatory chemokines and their receptors are the products of protooncogenes in many cancers including that of the prostate. This review will focus on the current biology of chemokines and chemokine receptors in prostate cancer. An understanding of this axis may enable researchers to develop targeted strategies for prostate cancer.
Collapse
Affiliation(s)
- Rajendra K Singh
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | | | | |
Collapse
|
23
|
Abstract
Background: Although the aetiology of prostate cancer remains unknown, we hypothesised that chronic bacterial insult has a major role in prostate carcinogenesis. Methods: Male C3H/HeOuJ mice, infected with phosphate-buffered saline or Escherichia coli bacteria, were killed at 5 days, or at 12 or 26 weeks. Harvested prostate tissues were evaluated for inflammatory responses and immunostained for neoplastic transformation markers. Results: All infected mice developed bacterial prostatitis. Control mice had no prostate infections or inflammation. Mice infected for 5 days showed foci of acute inflammation with infiltrating neutrophils and epithelial necrotic debris in the prostatic glandular lumen. All mice infected for 12 weeks had evidence of chronic inflammation with dense inflammatory infiltrates in the stroma. The prostatic epithelium showed varying degrees of atypical hyperplasia with increased epithelial cell layers and cytological atypia. At 26 weeks, the dysplastic changes were more pronounced and mimicked a prostatic intraepithelial neoplasia and high-grade dysplasia. Prostatic glands exhibiting reactive dysplasia had a stronger staining for oxidative DNA damage, increased epithelial cell proliferation, and a decrease in androgen receptor, GSTP1, p27Kip1, and PTEN expression, when compared with control prostate glands. Conclusion: These data demonstrate that chronic inflammation induces focal prostatic glandular atypia and suggest a potential linkage between inflammation and prostatic neoplasia.
Collapse
|
24
|
Mo Z, Gao Y, Cao Y, Gao F, Jian L. An updating meta-analysis of the GSTM1, GSTT1, and GSTP1 polymorphisms and prostate cancer: a HuGE review. Prostate 2009; 69:662-88. [PMID: 19143011 DOI: 10.1002/pros.20907] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
It has been postulated that individuals with GSTM1, GSTT1 deficiency and, GSTP1 (105Ile/Val transition) have increased susceptibility to carcinogens and are more likely to develop prostate cancer. In recent years, GST status has been extensively studied as a prostate cancer risk factor; however, the results are inconsistent. To re-examine this controversy, we have undertaken an updating meta-analysis of 29 studies with GSTM1 genotyping (4,564 prostate cancer cases and 5,464 controls), 22 studies with GSTT1 genotyping (3,837 cases and 4,552 controls), and 24 studies with GSTP1 genotyping (5,301 cases and 5,621 controls). The random effects odds ratio was 1.33 [95% confidence interval (95% CI): 1.15, 1.55; I(2) = 68.9%, P for heterogeneity = 0.00] for the GSTM1 null versus present genotype and 1.05 (95% CI: 0.86, 1.27; I(2) = 68.2%, P for heterogeneity = 0.00) for the GSTT1 null versus present genotype, and 1.06 (95% CI: 0.91, 1.24; I(2) = 71.5%, P for heterogeneity = 0.00) for the GSTP1-Val versus GSTP1-Ile allele. For GSTM1 polymorphism, similar results reached in Caucasians and Asians, with exception for Africans. No association between GSTT1 or GSTP1 polymorphisms and prostate cancer risk was detected in different racial. In conclusion, the major finding of our study suggested that GSTM1 polymorphism conferred an increasing risk of prostate cancer on a wide population basis, however, no relationship was found between GSTT1 and GSTP1 status and the risk of prostate cancer.
Collapse
Affiliation(s)
- Zengnan Mo
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China.
| | | | | | | | | |
Collapse
|
25
|
Takayama H, Nonomura N, Nishimura K, Oka D, Shiba M, Nakai Y, Nakayama M, Tsujimura A, Aozasa K, Okuyama A. Decreased immunostaining for macrophage scavenger receptor is associated with poor prognosis of prostate cancer. BJU Int 2008; 103:470-4. [PMID: 18778349 DOI: 10.1111/j.1464-410x.2008.08013.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE The aim of this study is to evaluate the expression of the macrophage scavenger receptor (MSR) in prostate needle biopsy specimens as a possible prognostic factor for prostate cancer. As MSR reportedly has a role in recognizing foreign pathogenic substances, MSR-positive inflammatory cells are often detected in solid tumours, and there is a correlation between the relative risk of prostate cancer and polymorphism of the MSR gene. PATIENTS AND METHODS MSR was evaluated by immunostaining in needle biopsies of the prostate from 135 patients who were confirmed to have prostate cancer. Among these men, 70 were treated by radical prostatectomy or by radiotherapy as definitive therapy; the other 65 were treated by hormonal therapy because of advanced disease or age. Needle-biopsy specimens were sectioned at 5 microm and immunostained with a monoclonal antibody against MSR. Six microscopic (x400) fields around the cancer foci were selected in each case for analysis. RESULTS The median number of MSR-positive cells (MSR count) in each case was 24. There was an inverse correlation between the MSR count and Gleason score and clinical stage. The MSR count was lower in patients with biochemical (prostate-specific antigen, PSA) failure than that in those with no PSA failure (P < 0.001). In all patients, the recurrence-free survival (RFS) rate was significantly higher in those with a high MSR count (> or =24) than that in those with low MSR count (<24, P < 0.001). Moreover, for patients treated by definitive or hormonal therapy, the RFS rates in those with a higher MSR count were higher than in those with a lower MSR count (P < 0.001 and 0.014, respectively). Cox multivariate analysis showed that the MSR count was a prognostic factor for prostate cancer in addition to extraprostatic extension and Gleason score (P = 0.002, 0.038 and 0.011, respectively). CONCLUSION The results of immunostaining of MSR in needle-biopsy specimens is a prognostic factor for prostate cancer.
Collapse
Affiliation(s)
- Hitoshi Takayama
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Katoh T, Yamano Y, Tsuji M, Watanabe M. Genetic polymorphisms of human cytosol glutathione S-transferases and prostate cancer. Pharmacogenomics 2008; 9:93-104. [PMID: 18154451 DOI: 10.2217/14622416.9.1.93] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Glutathione S-transferases (GSTs) are involved in the metabolism of a wide range of carcinogenic chemicals. In humans, cytosol GSTs are divided into eight classes: alpha (GSTA), mu (GSTM), pi (GSTP), theta (GSTT), tau (GSTZ), sigma (GSTS), omicron (GSTO) and kappa (GSTK). The allelic polymorphism of these enzymes is associated with variations in enzyme activity; hence, it may affect the concentration of activated carcinogenic chemicals in the body. In addition to the metabolism of chemical carcinogens, GSTs metabolize steroid hormones, compounds in the diet and other agents potentially involved in prostate carcinogenesis. Three genetic polymorphisms of GSTs, GSTM1*0 (null), GSTT1*0 (null) and GSTP1 A313G, have been well documented. No consistent associations between GSTM1, GSTT1 or GSTP1 genotypes and prostate cancer have been observed. Recent meta-analysis reports show that these polymorphisms of GSTM1, GSTT1 and GSTP1 are unlikely to be major determinants of susceptibility to prostate cancer.
Collapse
Affiliation(s)
- Takahiko Katoh
- Kumamoto University, Department of Public Health, Graduate school of Medical Sciences, Kumamoto 860-8556, Japan.
| | | | | | | |
Collapse
|
27
|
|
28
|
Hurt EM, Thomas SB, Peng B, Farrar WL. Molecular consequences of SOD2 expression in epigenetically silenced pancreatic carcinoma cell lines. Br J Cancer 2007; 97:1116-23. [PMID: 17895890 PMCID: PMC2360443 DOI: 10.1038/sj.bjc.6604000] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2007] [Revised: 08/14/2007] [Accepted: 08/29/2007] [Indexed: 02/06/2023] Open
Abstract
Manganese superoxide dismutase (SOD2) is an enzyme that catalyses the dismutation of superoxide in the mitochondria, leading to reduced levels of reactive oxygen species. Reduced expression levels of SOD2 have been shown to result in increased DNA damage and sod2 heterozygous mice have increased incidences of cancer. It has also been shown that SOD2 expression is lost in pancreatic cell lines, with reintroduction of SOD2 resulting in decreased rate of proliferation. The mechanism of decreased SOD2 expression in pancreatic carcinoma has not been previously determined. We demonstrate, through sodium bisulphite sequencing, that the sod2 locus is methylated in some pancreatic cell lines leading to a corresponding decrease in SOD2 expression. Methylation can be reversed by treatment with zebularine, a methyltransferase inhibitor, resulting in restored SOD2 expression. Furthermore, we demonstrate that sensitivity of pancreatic carcinoma cell lines to 2-methoxyestradiol correlates with SOD2 expression and SOD2 modulation can alter the sensitivity of these cells. Using both genomics and proteomics, we also identify molecular consequences of SOD2 expression in MIA-PaCa2 cells, including dephosphorylation of VEGFR2 and the identification of both SOD2-regulated genes and transcription factors with altered binding activity in response to SOD2 expression.
Collapse
Affiliation(s)
- E M Hurt
- Cancer Stem Cell Section, Laboratory of Cancer Prevention, Center for Cancer Research, National Cancer Institute at Frederick, National Institutes of Health, Frederick, MD 21702, USA
| | - S B Thomas
- Basic Research Program, SAIC-Frederick Inc., National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - B Peng
- School of Dental Science, University of Melbourne, Melbourne, Victoria, Australia
| | - W L Farrar
- Cancer Stem Cell Section, Laboratory of Cancer Prevention, Center for Cancer Research, National Cancer Institute at Frederick, National Institutes of Health, Frederick, MD 21702, USA
| |
Collapse
|
29
|
Waters DJ, Shen S, Xu H, Kengeri SS, Cooley DM, Chiang EC, Chen Y, Schlittler D, Oteham C, Combs GF, Glickman LT, Morris JS, Bostwick DG. Noninvasive Prediction of Prostatic DNA Damage by Oxidative Stress Challenge of Peripheral Blood Lymphocytes. Cancer Epidemiol Biomarkers Prev 2007; 16:1906-10. [PMID: 17855713 DOI: 10.1158/1055-9965.epi-07-0034] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
To move closer to the goal of individualized risk prediction for prostate cancer, we used an in vivo canine model to evaluate whether the susceptibility of peripheral blood lymphocytes (PBLs) to oxidative stress-induced DNA damage could identify those individuals with the highest prostatic DNA damage. This hypothesis was tested in a population of 69 elderly male beagle dogs after they had completed a 7-month randomized feeding trial to achieve the broad range of dietary selenium status observed in U.S. men. The alkaline Comet assay was used to directly compare the extent of DNA damage in PBLs with prostatic DNA damage in each dog. Using stepwise logistic regression, the sensitivity of PBLs to oxidative stress challenge with hydrogen peroxide (H(2)O(2)) predicted dogs in the highest tertile of prostatic DNA damage. Dogs with PBLs highly sensitive to H(2)O(2) were 7.6 times [95% confidence interval (95% CI), 1.5-38.3] more likely to have high prostatic DNA damage than those in the H(2)O(2)-resistant group. This risk stratification was observed in multivariate analysis that considered other factors that might influence DNA damage, such as age, toenail selenium concentration, and serum testosterone concentration. Our data show that the sensitivity of PBLs to oxidative stress challenge, but not endogenous DNA damage in PBLs, provides a noninvasive surrogate marker for prostatic DNA damage. These findings lend support to the concept that oxidative stress contributes to genotoxic damage, and that oxidative stress challenge may stratify men for prostate cancer risk.
Collapse
Affiliation(s)
- David J Waters
- Gerald P. Murphy Cancer Foundation, 3000 Kent Avenue, Suite E2-100, West Lafayette, IN 47906, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Meiers I, Shanks JH, Bostwick DG. Glutathione S-transferase pi (GSTP1) hypermethylation in prostate cancer: review 2007. Pathology 2007; 39:299-304. [PMID: 17558856 DOI: 10.1080/00313020701329906] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Prostatic carcinoma is characterised by the silencing of the pi-class glutathione S-transferase gene (GSTP1), which encodes a detoxifying enzyme. The silencing of GSTP1 results from aberrant methylation at the CpG island in the promoter-5' and occurs in the vast majority of cases of high-grade prostatic intraepithelial neoplasia (PIN) and prostate cancers. We review the potential novel role of GSTP1 and its related expression in prostate cancer. The loss of expression (silencing) of the GSTP1 gene is the most common (>90%) genetic alteration reported to date in prostate cancer. Quantitative methylation-specific PCR assays allow detection of GSTP1 methylation in prostate biopsies and may improve the sensitivity of cancer detection. Advances in the epigenetic characterisation of prostate cancer have enabled the development of DNA methylation assays that may soon be used in diagnostic testing of serum and tissue for prostate cancer. Inhibition of aberrant promoter methylation could theoretically prevent carcinogenesis.
Collapse
Affiliation(s)
- Isabelle Meiers
- Department of Pathology, Bostwick Laboratories, London, United Kingdom.
| | | | | |
Collapse
|
31
|
. HSAELA, . AZS, . AF. Impact of Whey Proteins on the Genotoxic Effects of Aflatoxins in Rats. ACTA ACUST UNITED AC 2007. [DOI: 10.3923/ijds.2007.126.137] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
32
|
Borowsky AD, Dingley KH, Ubick E, Turteltaub KW, Cardiff RD, Devere-White R. Inflammation and atrophy precede prostatic neoplasia in a PhIP-induced rat model. Neoplasia 2006; 8:708-15. [PMID: 16984728 PMCID: PMC1584295 DOI: 10.1593/neo.06373] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
2-amino-1-methyl-6-phenylimidazo(4,5-b)pyridine (PhIP) has been implicated as a major mutagenic heterocyclic amine in the human diet and is carcinogenic in the rat prostate. To validate PhIP-induced rat prostatic neoplasia as a model of human prostate cancer progression, we sought to study the earliest histologic and morphologic changes in the prostate and to follow progressive changes over time. We fed sixty-seven 5-week-old male Fischer F344 rats with PhIP (400 ppm) or control diets for 20 weeks, and then sacrificed animals for histomorphologic examination at the ages of 25, 45, and 65 weeks. Animals treated with PhIP showed significantly more inflammation (P = .002, > .001, and .016 for 25, 45, and 65 weeks, respectively) and atrophy (P = .003, > .001, and .006 for 25, 45, and 65 weeks, respectively) in their prostate glands relative to controls. Prostatic intraepithelial neoplasia (PIN) occurred only in PhIP-treated rats. PIN lesions arose in areas of glandular atrophy, most often in the ventral prostate. Atypical cells in areas of atrophy show loss of glutathione S-transferase pi immunostaining preceding the development of PIN. None of the animals in this study developed invasive carcinomas, differing from those in previous reports. Overall, these findings suggest that the pathogenesis of prostatic neoplasia in the PhIP-treated rat prostate proceeds from inflammation to postinflammatory proliferative atrophy to PIN.
Collapse
|
33
|
Abstract
The development of targeted therapies for prostate cancer has exploited various elements of prostate biology. The androgen-dependence of prostate cancer continues to be the focus for the development of new drugs and the analysis of details of the intermolecular interactions of the androgen receptor. Importantly, new applications of androgen ablation therapy have proven to have the greatest effect on cause-specific and overall survival during the last decade. Prostate epithelial cells express a number of tissue-specific proteins that have been the target either for antibody-directed therapies, in the case of prostate-specific membrane antigen, or target-activated therapies in the case of prostate-specific antigen, a serine protease. Prostate-specific proteins have also been targeted by the development of vaccines that have entered clinical trials. Humanized monoclonal antibodies and small molecules designed to inhibit oncogenic signalling pathways have been subjected to clinical trials in prostate cancer with limited success. The application of pathway inhibitors to prostate cancer therapy has been limited because no common dominant oncogenic mutation affecting signal kinase activation in prostate cancer has yet been identified. The interaction of signal kinase inhibitors with androgen ablation and with cytotoxic chemotherapy remains to be explored.
Collapse
Affiliation(s)
- Ekatherine Asatiani
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3800 Reservoir Road, NW Washington, DC 20007-2197, USA
| | | |
Collapse
|
34
|
Lockett KL, Hall MC, Clark PE, Chuang SC, Robinson B, Lin HY, Su LJ, Hu JJ. DNA damage levels in prostate cancer cases and controls. Carcinogenesis 2005; 27:1187-93. [PMID: 16364923 DOI: 10.1093/carcin/bgi288] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
This study used the alkaline Comet assay to evaluate whether basal or H2O2-induced DNA damage is associated with prostate cancer (CaP) risk. Using lymphocyte samples from 158 CaP cases and 128 controls, collected in an ongoing case-control study, our results showed that basal DNA damage did not differ between cases and controls. However, the H2O2-induced DNA damage level was significantly higher in incident cases (mean +/- SD; 6.61 +/- 4.43, n = 102) than controls (5.30 +/- 3.60, n = 128) or prevalent cases (4.47 +/- 3.19; n = 56). Incident cases with a positive smoking history had significantly higher H2O2-induced DNA damage than never-smokers (7.57 +/- 4.82 versus 4.52 +/- 2.40; P < 0.001). Above-median H2O2-induced DNA damage was associated with a 1.61-fold increase in CaP risk [95% confidence interval (CI) = 0.92-2.81], after adjustment for age, race, benign prostatic hyperplasia (BPH), smoking history and family history (FH). Using the lowest quartile of H2O2-induced DNA damage as the referent group, the adjusted ORs for the 25th, 50th and 75th quartiles were 0.90 (95% CI = 0.39-2.05), 1.06 (95% CI = 0.48-2.35) and 2.05 (95% CI = 0.96-4.37), respectively (P = 0.046, test for linear trend). The association between CaP and DNA damage was modified by age, smoking history, family history and body mass index. Our results suggest that DNA damage may be associated with CaP risk. However, larger case-control and follow-up studies are warranted to further evaluate the potential application of the alkaline Comet assay in CaP risk assessment and prevention.
Collapse
Affiliation(s)
- Kristin L Lockett
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Crowell JA. The chemopreventive agent development research program in the Division of Cancer Prevention of the US National Cancer Institute: an overview. Eur J Cancer 2005; 41:1889-910. [PMID: 16005206 DOI: 10.1016/j.ejca.2005.04.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2005] [Accepted: 04/25/2005] [Indexed: 02/06/2023]
Abstract
Chemoprevention is an innovative area of cancer research that focuses on the development of pharmacological, biological, and nutritional interventions to prevent, reverse, or delay carcinogenesis. Over the past two decades the Division of Cancer Prevention of the US National Cancer Institute has organized a research and development program to provide resources and infrastructure to the research community for the clinical evaluation of potential cancer preventive agents. This program now encompasses preclinical agent and molecular target identification, in vitro and in vivo screening, efficacy and intermediate endpoint testing, pharmacology and toxicology assessments, and finally chemical synthesis and manufacturing leading to Investigational New Drug applications and clinical studies. In this review, examples of agents currently in development, preclinical testing models, and phase 1 and 2 clinical studies are described. Continued commitment to cancer prevention will significantly reduce the economic and medical burden of cancer.
Collapse
Affiliation(s)
- James A Crowell
- Division of Cancer Prevention, National Cancer Institute, DHHS, Bethesda, MD 20892, USA
| |
Collapse
|
36
|
Jerónimo C, Henrique R, Hoque MO, Mambo E, Ribeiro FR, Varzim G, Oliveira J, Teixeira MR, Lopes C, Sidransky D. A quantitative promoter methylation profile of prostate cancer. Clin Cancer Res 2005; 10:8472-8. [PMID: 15623627 DOI: 10.1158/1078-0432.ccr-04-0894] [Citation(s) in RCA: 185] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Promoter hypermethylation is an alternative pathway for gene silencing in neoplastic cells and a promising cancer detection marker. Although quantitative methylation-specific PCR (QMSP) of the GSTP1 promoter has demonstrated near perfect specificity for cancer detection in prostate biopsies, we postulated that identification and characterization of additional methylation markers might further improve its high (80-90%) sensitivity. EXPERIMENTAL DESIGN We surveyed nine gene promoters (GSTP1, MGMT, p14/ARF, p16/CDKN2A, RASSF1A, APC, TIMP3, S100A2, and CRBP1) by QMSP in tissue DNA from 118 prostate carcinomas, 38 paired high-grade prostatic intraepithelial neoplasias (HGPIN), and 30 benign prostatic hyperplasias (BPH). The methylation levels were calculated and were correlated with clinical and pathologic indicators. RESULTS Only the methylation frequencies of GSTP1 and APC were significantly higher in prostate carcinoma compared with BPH (P < 0.001). Methylation levels of GSTP1, APC, RASSF1A, and CRBP1, differed significantly between prostate carcinoma and HGPIN, and/or HGPIN or BPH (P < 0.0001). With QMSP and empirically defined cutoff values, the combined use of GSTP1 and APC demonstrated a theoretical sensitivity of 98.3% for prostate carcinoma, with 100% specificity. Methylation levels were found to correlate with tumor grade (GSTP1 and APC) and stage (GSTP1, RASSF1A, and APC). CONCLUSIONS Our data demonstrate the existence of a progressive increase of promoter methylation levels of several cancer-related genes in prostate carcinogenesis, providing additional markers to augment molecular detection of prostate carcinoma. Because methylation levels of GSTP1, APC, and RASSF1A are associated with advanced grade and stage, QMSP might augment the pathologic indicators currently used to predict tumor aggressiveness.
Collapse
Affiliation(s)
- Carmen Jerónimo
- Department of Otolaryngology-Head and Neck Surgery, Head and Neck Cancer Research Division, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Gastric cancer is believed to result in part from the accumulation of multiple genetic alterations leading to oncogene overexpression and tumor suppressor loss. Epigenetic alterations as a distinct and crucial mechanism to silence a variety of methylated tissue-specific and imprinted genes, have been extensively studied in gastric carcinoma and play important roles in gastric carcinogenesis. This review will briefly discuss the basic aspects of DNA methylation and CpG island methylation, in particular the epigenetic alterations of certain critical genes implicated in gastric carcinogenesis and its relevance of clinical implications.
Collapse
Affiliation(s)
- In Seon Choi
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | | |
Collapse
|
38
|
Mai A, Massa S, Rotili D, Cerbara I, Valente S, Pezzi R, Simeoni S, Ragno R. Histone deacetylation in epigenetics: an attractive target for anticancer therapy. Med Res Rev 2005; 25:261-309. [PMID: 15717297 DOI: 10.1002/med.20024] [Citation(s) in RCA: 256] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The reversible histone acetylation and deacetylation are epigenetic phenomena that play critical roles in the modulation of chromatin topology and the regulation of gene expression. Aberrant transcription due to altered expression or mutation of genes that encode histone acetyltransferase (HAT) or histone deacetylase (HDAC) enzymes or their binding partners, has been clearly linked to carcinogenesis. The histone deacetylase inhibitors are a new promising class of anticancer agents (some of which in clinical trials), that inhibit the proliferation of tumor cells in culture and in vivo by inducing cell-cycle arrest, terminal differentiation, and/or apoptosis. This report reviews the chemistry and the biology of HDACs and HDAC inhibitors, laying particular emphasis on agents actually in clinical trials for cancer therapy and on new potential anticancer lead compounds more selective and less toxic.
Collapse
Affiliation(s)
- Antonello Mai
- Istituto Pasteur, Fondazione Cenci-Bolognetti, Dipartimento di Studi Farmaceutici, Università degli Studi di Roma La Sapienza, P.le A. Moro 5, 00185 Roma, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Chan QKY, Khoo US, Chan KYK, Ngan HYS, Li SS, Chiu PM, Man LS, Ip PPC, Xue WC, Cheung ANY. Promoter methylation and differential expression of pi-class glutathione S-transferase in endometrial carcinoma. J Mol Diagn 2005; 7:8-16. [PMID: 15681469 PMCID: PMC1867507 DOI: 10.1016/s1525-1578(10)60003-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Pi-class glutathione S-transferase (GSTP1), located on chromosome 11q13, codes for a phase II metabolic enzyme that detoxifies reactive electrophilic intermediates. The protein also interacts with steroid hormones in the human body. The role of GSTP1 in endometrial carcinoma has not been reported. In this study, we aimed at determining the expression of GSTP1 in relation to the epigenetic and genetic changes of the gene in endometrial carcinoma. The GSTP1 protein and mRNA expression was assessed by immunohistochemistry on tissue microarray and quantitative real-time reverse transcriptase-polymerase chain reaction, respectively. Its methylation status was studied by methylation-specific polymerase chain reaction and bisulfite sequencing. Possible mutations in coding region of GSTP1 were assessed by cDNA sequencing. Ninety-seven cases of endometrial carcinoma with available tissue blocks and clinical data were studied. Our results showed that 68.0% (66 of 97) of the cases showed reduced protein expression while 64% (16 of 25) showed reduced mRNA expression; 30.9% (30 of 97) of the cases demonstrated methylated alleles in at least one of the six methylation-specific polymerase chain reaction reactions. The methylation status significantly correlated with reduced protein expression (P = 0.008) and reduced mRNA expression (P = 0.003). Methylation at non-CpG sites including CpCpG trinucleotides and CpT dinucleotides were also observed. cDNA sequencing did not reveal genetic alterations in coding region of the gene. The extent of myometrial invasion was found to be significantly correlated with both the methylation status (P = 0.009) and the protein expression (P = 0.036) of the GSTP1 gene. We postulated that hypermethylation of the GSTP1 gene promoter region may act as a dynamic regulation mechanism contributing to reduced GSTP1 expression, which is associated with myometrial invasion potential of the endometrial carcinoma.
Collapse
Affiliation(s)
- Queeny K Y Chan
- Department of Pathology, Queen Mary Hospital, The University of Hong Kong, Pokfulam Rd., Hong Kong
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Antognelli C, Mearini L, Talesa VN, Giannantoni A, Mearini E. Association of CYP17, GSTP1, and PON1 polymorphisms with the risk of prostate cancer. Prostate 2005; 63:240-51. [PMID: 15538743 DOI: 10.1002/pros.20184] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Dietary factors, life-style as well as environmental conditions may contribute to the risk of prostate tumor together with genetic susceptibility, that may be an important factor in determining who is more likely to develop prostate malignancy. We have undertaken a case-control study in order to elucidate the association between polymorphisms in some metabolizing genes with the risk of prostate cancer (PCa). METHODS Polymorphisms of three xenobiotic genes (CYP17, GSTP1, PON1) were characterized in 384 patients with untreated PCa and 360 age-matched control patients with benign prostatic hyperplasia (BPH). All polymorphisms were investigated by PCR/RFLP methods using DNA from lymphocytes. RESULTS We found that men with the CYP17/A1A1-A1A2 genotypes, GSTP1/IleVal genotype, PON192/QR and PON55/LM-MM genotypes had a significantly higher risk of PCa compared with the others genotypes. CONCLUSIONS The three polymorphisms appear to be common genetic traits that are associated with an increased risk for PCa: the analysis of them all in each single case may be a predictable factor, particularly among groups exposed to PCa-related carcinogens.
Collapse
Affiliation(s)
- Cinzia Antognelli
- Experimental Medicine Department, University of Perugia, Perugia, Italy
| | | | | | | | | |
Collapse
|
41
|
Bernardini S, Miano R, Iori R, Finazzi-Agrò E, Palmieri G, Ballerini S, Angeloni C, Orlandi A, Bellincampi L, Cortese C, Federici G. Hypermethylation of the CpG islands in the promoter region of the GSTP1 gene in prostate cancer: a useful diagnostic and prognostic marker? Clin Chim Acta 2005; 350:181-8. [PMID: 15530476 DOI: 10.1016/j.cccn.2004.07.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2004] [Revised: 07/23/2004] [Accepted: 07/23/2004] [Indexed: 11/20/2022]
Abstract
BACKGROUND Recently, many studies have focused on the potential diagnostic value of the promoter hypermethylation of the GSTP1 gene in prostate cancer. METHOD A total of 144 patients, undergoing eight-core prostatic biopsies for a clinically suspected prostate cancer, was analyzed. Two different tissue samples were collected from the same area of the prostate and then divided for both genomic DNA extraction and pathological examination. In order to perform molecular analysis, prostatic tissue samples were digested with the methylation-sensitive restriction enzyme HpaII and then amplified by conventional polymerase chain reaction (PCR). RESULTS Prostate cancer was diagnosed in 42/144 patients, and promoter hypermethylation of GSTP1 gene was detected in 31/42 of prostate cancer (sensitivity=74%) and in 2/102 of negative specimens (specificity=98%). A significant association between GSTP1 promoter hypermethylation both with a Gleason score >or=7 (Fisher's exact P=0.01) and the presence of Gleason grade 4 and/or grade 5 (Fisher's exact P=0.03) was found. CONCLUSION Promoter hypermethylation of the GSTP1 gene is a highly specific--but not a very sensitive--marker of prostate cancer. Our data showed a significant association between the methylation status of the GSTP1 gene and Gleason score and grade, suggesting a potential prognostic value of this epigenetic DNA alteration.
Collapse
Affiliation(s)
- Sergio Bernardini
- Department of Internal Medicine, Policlinico Tor Vergata, University of Tor Vergata, Rome, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Bastian PJ, Yegnasubramanian S, Palapattu GS, Rogers CG, Lin X, De Marzo AM, Nelson WG. Molecular biomarker in prostate cancer: the role of CpG island hypermethylation. Eur Urol 2005; 46:698-708. [PMID: 15548435 DOI: 10.1016/j.eururo.2004.07.022] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2004] [Indexed: 12/31/2022]
Abstract
CpG island hypermethylation may be one of the earliest somatic genome alterations to occur during the development of multiple cancers. Recently, aberrant methylation patterns for different tumors have been reported. We present a comprehensive review of the literature describing the role of CpG island hypermethylation of DNA from prostatic tissue and bodily fluids from men with prostate cancer. We reviewed the literature to evaluate CpG island hypermethylation in tissue and bodily fluids of men with primary and metastatic prostate cancer. Additionally, we reviewed the literature with respect to CpG island hypermethylation patterns in other urological malignancies. Using modern analytic methods, CpG island hypermethylation detection can be achieved. In men with prostate cancer, correlations between specific gene regulatory region hypermethylation analyses and Gleason score, pathologic stage and tumor recurrence have been demonstrated. CpG island hypermethylation may serve as a useful molecular biomarker for the detection and diagnosis of patients with prostate cancer.
Collapse
Affiliation(s)
- Patrick J Bastian
- The James Buchanan Brady Urological Institute, Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231-1000, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Lockett KL, Hall MC, Xu J, Zheng SL, Berwick M, Chuang SC, Clark PE, Cramer SD, Lohman K, Hu JJ. The ADPRT V762A genetic variant contributes to prostate cancer susceptibility and deficient enzyme function. Cancer Res 2004; 64:6344-8. [PMID: 15342424 DOI: 10.1158/0008-5472.can-04-0338] [Citation(s) in RCA: 138] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The ADP-ribosyltransferase (ADPRT) gene encodes a zinc-finger DNA-binding protein, poly(ADP-ribose) polymerase-1 (PARP-1), that modifies various nuclear proteins by poly(ADP-ribosyl)ation and functions as a key enzyme in the base excision repair pathway. We have conducted two studies to test whether an amino acid substitution variant, ADPRT V762A (T2444C), is associated with prostate cancer (CaP) risk and decreased enzyme function. The first study used genomic DNA samples from an ongoing, clinic-based case-control study (488 cases and 524 controls) to show that a higher percentage of the CaP cases carried the ADPRT 762 AA genotype than controls (4% versus 2%). In Caucasians, the AA genotype was significantly associated with increased CaP risk [odds ratio (OR), 2.65; 95% confidence interval (CI), 1.08-6.49], and the VA genotype was associated with a slight but not significantly increased CaP risk (OR, 1.18; 95% CI, 0.85-1.64) using VV as the referent group after adjustment for age, benign prostatic hyperplasia, and family history. Furthermore, this association was stronger in younger (<65) men (OR, 4.77; 95% CI, 1.01-22.44) than older (> or =65) men (OR, 1.78; 95% CI, 0.55-5.82). The second study used freshly isolated peripheral lymphocytes from 354 cancer-free subjects to demonstrate that the ADPRT 762 A allele contributed to significantly lower adenosine diphosphate ribosyl transferase (ADPRT)/PARP-1 activities in response to H2O2 in a gene dosage-dependent manner (P < 0.0001, test for linear trend). The PARP-1 activities (mean +/- SD dpm/10(6) cells) were 18,554 +/- 9,070 (n=257), 14,847 +/- 7,082 (n=86), and 12,155 +/- 6,334 (n=11) for VV, VA, and AA genotypes, respectively. This study is the first to provide evidence that the ADPRT V762A-genetic variant contributes to CaP susceptibility and altered ADPRT/PARP-1 enzyme function in response to oxidative damage.
Collapse
Affiliation(s)
- Kristin L Lockett
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, 27157, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Beer TM, Evans AJ, Hough KM, Lowe BA, McWilliams JE, Henner WD. Polymorphisms of GSTP1 and related genes and prostate cancer risk. Prostate Cancer Prostatic Dis 2004; 5:22-7. [PMID: 15195126 DOI: 10.1038/sj.pcan.4500549] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2001] [Accepted: 08/28/2001] [Indexed: 11/09/2022]
Abstract
Glutathione S-transferase P1 (GSTP1) is markedly downregulated in prostate cancer and prostatic intraepithelial neoplasia compared to normal prostate tissue. Downregulation of GSTP1 may, therefore, be an early event in prostate carcinogenesis. An A-->G polymorphism at nucleotide 313 results in an amino acid substitution (Ile105Val) in the substrate binding site of GSTP1 and reduces catalytic activity of GSTP1. In a study of 36 prostate cancer patients, Harries et al. reported that the Ile/Ile genotype is associated with a decreased risk of prostate cancer (odds ratio 0.4 (0.17-0.82)). We sought to confirm this finding and to examine the impact of this polymorphism together with several related polymorphisms implicated as risk factors for carcinogen-associated malignancies. One hundred and seventeen patients with prostate adenocarcinoma and 183 population-based controls were recruited to this case-control study. Genotyping of the GSTP1 (Ile105Val), GSTM1 (null), GSTT1 (null) and CYP1A1 (Ile462Val) genes was performed using polymerase chain reaction (PCR) based techniques on DNA prepared from peripheral blood. A questionnaire was used to collect demographic information from each subject. Cases were significantly older (P<0.0001) and had significantly greater family history of prostate cancer (P<0.0001), confirming known risk factors for this disease. By chi(2) analysis, none of the genotype distributions varied among cases and controls. Using a logistic regression model to control for known risk factors we were also unable to demonstrate a significant association with prostate cancer for any of the polymorphisms tested. This population fails to identify a relationship between the above polymorphisms and prostate adenocarcinoma.
Collapse
Affiliation(s)
- T M Beer
- Division of Hematology and Medical Oncology, Oregon Health Sciences University, Oregon, USA.
| | | | | | | | | | | |
Collapse
|
45
|
De Marzo AM, DeWeese TL, Platz EA, Meeker AK, Nakayama M, Epstein JI, Isaacs WB, Nelson WG. Pathological and molecular mechanisms of prostate carcinogenesis: implications for diagnosis, detection, prevention, and treatment. J Cell Biochem 2004; 91:459-77. [PMID: 14755677 DOI: 10.1002/jcb.10747] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Prostate cancer is an increasing threat throughout the world. As a result of a demographic shift in population, the number of men at risk for developing prostate cancer is growing rapidly. For 2002, an estimated 189,000 prostate cancer cases were diagnosed in the U.S., accompanied by an estimated 30,200 prostate cancer deaths [Jemal et al., 2002]. Most prostate cancer is now diagnosed in men who were biopsied as a result of an elevated serum PSA (>4 ng/ml) level detected following routine screening. Autopsy studies [Breslow et al., 1977; Yatani et al., 1982; Sakr et al., 1993], and the recent results of the Prostate Cancer Prevention Trial (PCPT) [Thompson et al., 2003], a large scale clinical trial where all men entered the trial without an elevated PSA (<3 ng/ml) were subsequently biopsied, indicate the prevalence of histologic prostate cancer is much higher than anticipated by PSA screening. Environmental factors, such as diet and lifestyle, have long been recognized contributors to the development of prostate cancer. Recent studies of the molecular alterations in prostate cancer cells have begun to provide clues as to how prostate cancer may arise and progress. For example, while inflammation in the prostate has been suggested previously as a contributor to prostate cancer development [Gardner and Bennett, 1992; Platz, 1998; De Marzo et al., 1999; Nelson et al., 2003], research regarding the genetic and pathological aspects of prostate inflammation has only recently begun to receive attention. Here, we review the subject of inflammation and prostate cancer as part of a "chronic epithelial injury" hypothesis of prostate carcinogenesis, and the somatic genome and phenotypic changes characteristic of prostate cancer cells. We also present the implications of these changes for prostate cancer diagnosis, detection, prevention, and treatment.
Collapse
Affiliation(s)
- Angelo M De Marzo
- Department of Oncology, The Johns Hopkins University School of Medicine, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21231-1000, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Chen JZ, Kadlubar FF. Mitochondrial mutagenesis and oxidative stress in human prostate cancer. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2004; 22:1-12. [PMID: 15845219 DOI: 10.1081/gnc-120037931] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Prostate cancer is the most common cancer diagnosed in men in the United States, but the primary cause and the molecular events leading to prostate carcinogenesis are poorly understood. Using the approach of laser capture microdissection, we revealed extensive somatic mitochondrial DNA (mtDNA) mutations in prostatic neoplastic lesions. Inspection of the lesion associated mutations not only provided new insights into the genetics of prostate cancer, but also revealed new patterns of mtDNA mutation in prostate carcinogenesis. Further analysis on a high frequency of multiple mutational events observed in the same neoplastic lesion revealed an unusually rapid process in mitochondrial mutagenesis, suggesting a new process of mitochondrial hyper-mutagenesis in cancer cells, likely mediated by cellular oxidative stress. Thus, active mitochondrial mutagenesis in prostate cancer suggests a prominent role of increased cellular oxidative stress in neoplastic transformation and the increased susceptibility of neoplastic cells to oxidative damage.
Collapse
Affiliation(s)
- Junjian Z Chen
- Division of Molecular Epidemiology, National Center for Toxicological Research, Jefferson, Arkansas 72079, USA.
| | | |
Collapse
|
47
|
Abstract
The diagnosis of prostatic adenocarcinoma, especially when present in small amounts, is often challenging. Before making a diagnosis of carcinoma, it is prudent for the pathologist to consider the various benign patterns and processes that can simulate prostatic adenocarcinoma. A useful method of classifying benign mimickers is in relationship to the major growth patterns depicted in the classical Gleason diagram. The four major patterns are small gland, large gland, fused gland and solid. Most mimickers fit within the small gland category and the most common ones giving rise to false-positive cancer diagnosis are atrophy, post-atrophic hyperplasia, atypical adenomatous hyperplasia and seminal vesicle-type tissue. A number of other histoanatomic structures such as Cowper's gland, verumontanum mucosal glands, mesonephric glands and paraganglionic tissue may be confused with adenocarcinoma. Additionally, metaplastic and hyperplastic processes within the prostate may be confused with adenocarcinoma. Furthermore, inflammatory processes including granulomatous prostatitis, xanthogranulomatous prostatitis and malakoplakia may simulate high-grade adenocarcinoma. Atypical adenomatous hyperplasia (adenosis), a putative precursor of transition zone adenocarcinoma, has overlapping features with low-grade adenocarcinoma and may cause problems in differential diagnosis, especially in the needle biopsy setting. The pathologist's awareness of the vast array of benign mimickers is important in the systematic approach to the diagnosis of prostatic adenocarcinoma. Knowledge of these patterns on routine microscopy coupled with the prudent use of immunohistochemistry will lead to a correct diagnosis and avert a false-positive cancer interpretation.
Collapse
Affiliation(s)
- John R Srigley
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
48
|
Abstract
PURPOSE We provide an overview of some of the basic, clinical and epidemiological research that has been conducted to investigate the potential role of chronic inflammation in prostate carcinogenesis and to provide direction for future research on this hypothesis. MATERIALS AND METHODS We reviewed the literature on this topic. RESULTS Chronic inflammation has long been linked to cancers with an infectious etiology, such as stomach, liver and colon cancer, in patients with inflammatory bowel disease. Whether intraprostatic inflammation contributes to prostate carcinogenesis is unknown. Inflammation is frequently present in prostate biopsies, radical prostatectomy specimens and tissue resected for treatment of benign prostatic hyperplasia. Also, inflammatory infiltrates are often found in and around foci of atrophy that are characterized by an increased proliferative index. These foci, called proliferative inflammatory atrophy, may be precursors of early prostate cancer or may indicate an intraprostatic environment favorable to cancer development. Epidemiological studies have indirectly examined the role of chronic inflammation in prostate carcinogenesis through studies of pro-inflammatory and anti-inflammatory factors. When taken together studies of sexually transmitted infections, clinical prostatitis, and genetic and circulating markers of inflammation and response to infection hint at a link between chronic intraprostatic inflammation and prostate cancer. CONCLUSIONS Additional well-designed basic, clinical and epidemiological studies are needed to resolve questions about the role of chronic inflammation in prostate carcinogenesis and to determine if intraprostatic inflammation is a rational target for chemoprevention.
Collapse
Affiliation(s)
- Elizabeth A Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, The Brady Urological Institute, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland 21205, USA.
| | | |
Collapse
|
49
|
Lucia MS, Torkko KC. Inflammation as a target for prostate cancer chemoprevention: pathological and laboratory rationale. J Urol 2004; 171:S30-4; discussion S35. [PMID: 14713750 DOI: 10.1097/01.ju.0000108142.53241.47] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE We review the literature addressing a potential causal role for chronic or recurrent inflammation or infection in the development of prostate cancer. MATERIALS AND METHODS A literature search was conducted using MEDLINE to identify articles on chronic inflammation as a risk factor for cancer, particularly prostate cancer. RESULTS A causal role for chronic or recurrent inflammation or infection in the development of prostate cancer has yet to be proven. Inflammation may contribute to carcinogenesis by 1 or more of several potentially interrelated mechanisms, including 1) the elaboration of cytokines and growth factors that favor tumor cell growth, 2) induction of cyclooxygenase-2 in macrophages and epithelial cells, and 3) generation of mutagenic reactive oxygen and nitrogen species. Chronic inflammation in the form of stromal and epithelial infiltrates of lymphocytes and histiocytes is extremely common in the peripheral zone of the prostate where most cancers arise. Although differences in histology and terminology exist for these inflammatory and atrophic lesions, as a group they often display evidence of epithelial proliferation. Heterogeneous expression of the GSTP1 gene in such lesions has been proposed as evidence for susceptibility to oxidative damage, thereby providing fertile ground for carcinogenesis. CONCLUSIONS Although the cumulative evidence demonstrates that chronic inflammation may be a legitimate target for chemopreventive efforts, more study is needed to prove its etiological role in prostate cancer.
Collapse
Affiliation(s)
- M Scott Lucia
- Department of Pathology and Preventive Medicine and Biometrics, Unibersity of Colorado Health Sciences Center, Denver, Colorado 80262, USA.
| | | |
Collapse
|
50
|
Gonzalgo ML, Nakayama M, Lee SM, De Marzo AM, Nelson WG. Detection of GSTP1 methylation in prostatic secretions using combinatorial MSP analysis. Urology 2004; 63:414-8. [PMID: 14972513 DOI: 10.1016/j.urology.2003.08.039] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2003] [Accepted: 08/29/2003] [Indexed: 12/31/2022]
Abstract
OBJECTIVES To evaluate the utility of methylation-specific polymerase chain reaction analysis of the pi-class glutathione-S-transferase (GSTP1) gene promoter in prostatic secretions for cancer detection and prognostication. METHODS Prostatic secretions were obtained from a total of 100 radical prostatectomy specimens immediately after surgical extirpation. GSTP1 promoter methylation was assessed by methylation-specific polymerase chain reaction analysis using two different primer sets. Correlations between GSTP1 promoter methylation and clinical and pathologic variables were examined. RESULTS The sensitivity for detection of GSTP1 methylation in prostatic secretions from men with clinically localized prostate cancer using two different primer sets was 76% and 54%. Methylation of the GSTP1 promoter was detected by both primer sets in 44% and by at least one primer set in 86% of the prostatic secretion specimens. The degree of methylation detected in the prostatic secretions was associated with the extent of cancer (predominant involvement of one or both sides of the gland; P = 0.02) and increasing age (P = 0.009). CONCLUSIONS Genomic DNA with GSTP1 promoter methylation can be detected in prostatic secretion specimens from the great majority of men with localized prostate cancer. Assays of GSTP1 promoter methylation in prostatic massage fluid or ejaculate may therefore serve as useful adjuncts to existing methods for prostate cancer screening and prognostication.
Collapse
Affiliation(s)
- Mark L Gonzalgo
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | | | | | | | | |
Collapse
|