1
|
Zhang J, Liu C, Luo W, Sun B. Role of SIRT7 in Prostate Cancer Progression: New Insight Into Potential Therapeutic Target. Cancer Med 2025; 14:e70786. [PMID: 40165597 PMCID: PMC11959159 DOI: 10.1002/cam4.70786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/20/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
Prostate cancer (PCa) is the second most common cancer in men worldwide, and understanding its molecular mechanisms is crucial for developing effective treatment strategies. SIRT7, a NAD+-dependent histone deacetylase, has emerged as a key regulator in PCa progression due to its roles in chromatin remodeling, DNA repair, and transcriptional regulation. Analysis of 492 PCa samples from The Cancer Genome Atlas (TCGA) via cBioPortal revealed that high SIRT7 expression is associated with poor prognosis in PCa patients. Mechanistically, SIRT7 deacetylates histone H3 at lysine 18 (H3K18Ac), a marker associated with aggressive tumors, suppressing tumor suppressor genes and promoting cancer cell proliferation and survival. Epithelial-mesenchymal transition (EMT) is a cellular biological process in which epithelial cells undergo specific molecular and morphological changes to transform into cells with characteristics of mesenchymal cells. SIRT7 also regulates EMT, and inhibiting SIRT7 in PCa cell lines reduces cell migration and invasion, highlighting its potential as a therapeutic target. In summary, the clinical significance of SIRT7 expression in PCa requires further research to elucidate its mechanisms. Developing specific inhibitors targeting SIRT7's deacetylase activity is a promising therapeutic strategy. SIRT7 plays a crucial role in regulating biological processes such as cell proliferation, cell cycle, and apoptosis in PCa through its epigenetic control of gene expression and maintenance of genomic stability. Therefore, SIRT7 may be a potential therapeutic target for PCa, and its expression could have prognostic value for PCa patients, providing important guidance for clinical monitoring and diagnosis by physicians.
Collapse
Affiliation(s)
- Jiale Zhang
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory DiseaseGuangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
- Guangzhou LaboratoryGuangzhouChina
| | - Chenxin Liu
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory DiseaseGuangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
- Guangzhou LaboratoryGuangzhouChina
| | - Wenting Luo
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory DiseaseGuangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
- Guangzhou LaboratoryGuangzhouChina
| | - Baoqing Sun
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory DiseaseGuangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
- Guangzhou LaboratoryGuangzhouChina
| |
Collapse
|
2
|
Guz J, Zarakowska E, Mijewski P, Wasilow A, Lesniewski F, Foksinski M, Brzoszczyk B, Jarzemski P, Gackowski D, Olinski R. Epigenetic DNA modifications and vitamin C in prostate cancer and benign prostatic hyperplasia: Exploring similarities, disparities, and pathogenic implications. Neoplasia 2024; 58:101079. [PMID: 39471555 PMCID: PMC11550371 DOI: 10.1016/j.neo.2024.101079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/23/2024] [Accepted: 10/21/2024] [Indexed: 11/01/2024]
Abstract
OBJECTIVES Benign Prostatic Hyperplasia (BPH) and Prostate Cancer (PC) are very common pathologies among aging men. Both disorders involve aberrant cell division and differentiation within the prostate gland. However, the direct link between these two disorders still remains controversial. A plethora of works have demonstrated that inflammation is a major causative factor in both pathologies. Another key factor involved in PC development is DNA methylation and hydroxymethylation. METHODS A broad spectrum of parameters, including epigenetic DNA modifications and 8-oxo-7,8-dihydro-2'-deoxyguanosine, was analyzed by two-dimensional ultraperformance liquid chromatography with tandem mass spectrometry in tissues of BPH, PC, and marginal one, as well as in leukocytes of the patients and the control group. In the same material, the expression of TETs and TDG genes was measured using RT-qPCR. Additionally, vitamin C was quantified in the blood plasma and within cells (leukocytes and prostate tissues). RESULTS Unique patterns of DNA modifications and intracellular vitamin C (iVC) in BPH and PC tissues, as well as in leukocytes, were found in comparison with control samples. The majority of the alterations were more pronounced in leukocytes than in the prostate tissues. CONCLUSIONS Characteristic DNA methylation/hydroxymethylation and iVC profiles have been observed in both PC and BPH, suggesting potential shared molecular pathways between the two conditions. As a fraction of leukocytes may be recruited to the pathological tissues and can migrate back into the circulation/blood, the observed alterations in leukocytes may reflect dynamic changes associated with the PC development, suggesting their potential utility as early markers of prostate cancer development.
Collapse
Affiliation(s)
- Jolanta Guz
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz 85-092, Poland
| | - Ewelina Zarakowska
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz 85-092, Poland
| | - Pawel Mijewski
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz 85-092, Poland
| | - Aleksandra Wasilow
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz 85-092, Poland
| | - Fabian Lesniewski
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz 85-092, Poland
| | - Marek Foksinski
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz 85-092, Poland
| | - Bartosz Brzoszczyk
- Department of Urology, Jan Biziel University Hospital, Bydgoszcz 85-168; Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Poland
| | - Piotr Jarzemski
- Department of Urology, Jan Biziel University Hospital, Bydgoszcz 85-168; Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Poland
| | - Daniel Gackowski
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz 85-092, Poland.
| | - Ryszard Olinski
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz 85-092, Poland.
| |
Collapse
|
3
|
Terrazzan A, Vanini R, Ancona P, Bianchi N, Taccioli C, Aguiari G. State-of-the-art in transposable element modulation affected by drugs in malignant prostatic cancer cells. J Cell Biochem 2024; 125:e30557. [PMID: 38501160 DOI: 10.1002/jcb.30557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 02/14/2024] [Accepted: 03/07/2024] [Indexed: 03/20/2024]
Abstract
Over recent years, the investigation of transposable elements (TEs) has granted researchers a deeper comprehension of their characteristics and functions, particularly regarding their significance in the mechanisms contributing to cancer development. This manuscript focuses on prostate carcinoma cell lines and offers a comprehensive review intended to scrutinize the associations and interactions between TEs and genes, as well as their response to treatment using various chemical drugs, emphasizing their involvement in cancer progression. We assembled a compendium of articles retrieved from the PubMed database to construct networks demonstrating correlations with genes and pharmaceuticals. In doing so, we linked the transposition of certain TE types to the expression of specific transcripts directly implicated in carcinogenesis. Additionally, we underline that treatment employing different drugs revealed unique patterns of TE reactivation. Our hypothesis gathers the current understanding and guides research toward evidence-based investigations, emphasizing the association between antiviral drugs, chemotherapy, and the reduced expression of TEs in patients affected by prostate cancer.
Collapse
Affiliation(s)
- Anna Terrazzan
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Laboratory for Advanced Therapy Technologies (LTTA), University of Ferrara, Ferrara, Italy
| | - Riccardo Vanini
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Pietro Ancona
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Nicoletta Bianchi
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Cristian Taccioli
- Department of Animal Medicine, Production and Health (MAPS), University of Padua, Padua, Italy
| | - Gianluca Aguiari
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| |
Collapse
|
4
|
Sun D, Guo J, Liang W, Chen Y, Wei S, Li A, Wang L, Chen X. Histone methyltransferase SUV39H2 regulates apoptosis and chemosensitivity in prostate cancer through AKT/FOXO signaling pathway. Med Oncol 2024; 41:44. [PMID: 38170382 DOI: 10.1007/s12032-023-02252-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/08/2023] [Indexed: 01/05/2024]
Abstract
Prostate cancer (PCa) is one of the most common malignant tumors that exhibit both chemoresistance and recurrence. SUV39H2 is highly expressed in many types of human tumors, but its role in the development and progression of PCa has never been clarified. The aim of this study is to elucidate the role of SUV39H2 in the development and progression of PCa, its association with the AKT/FOXO signaling pathway, and its potential implications for PCa diagnosis and treatment. SUV39H2 expression was analyzed in The Cancer Genome Atlas (TCGA) and genotype tissue expression pan-cancer data. The TCGA database was evaluated for SUV39H2 enrichment and its correlation to immune cell infiltration. SUV39H2 levels in PCa tissues and control tissues were determined in 30 patients using qPCR and IHC. Clinical relevance was assessed via The Cancer Genome Atlas (TCGA). In vitro assessments including colony formation assays, Western Blot analysis, CCK-8 assays, and flow cytometry were utilized to establish SUV39H2's contribution to PCa cell growth. The influence of SUV39H2 on PC3 and DU145 cell proliferation was assessed through a cell line-derived xenograft model. Sphere formation assays and qPCR were employed to delineate SUV39H2's role in PCa stemness and chemosensitivity. In vitro macrophage polarization assays provided insights into SUV39H2's association with M2 macrophages, while enrichment analysis shed light on its role in FOXO signaling. PCa tissues expressed higher levels of SUV39H2 than normal tissues. By knocking down SUV39H2, PCa cells were made more chemosensitive to docetaxel and cell proliferation and stemness were inhibited. Additionally, SUV39H2 knockdown significantly inhibited in vivo PCa cell growth and inhibited the polarization of macrophages. Furthermore, SUV39H2 was found to regulate AKT/FOXO signaling by increasing Akt and FOXO3a phosphorylation. Our findings highlight SUV39H2's role in PCa cell apoptosis and chemosensitivity mainly by regulating the AKT/FOXO signaling pathway and suggest that SUV39H2 could be a potential target for PCa diagnosis and treatment.
Collapse
Affiliation(s)
- Donglin Sun
- Department of Urology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518100, China
| | - Jing Guo
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510000, China
| | - Weifei Liang
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511500, Guangdong, China
| | - Yangxiao Chen
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510000, China
| | - Shuqi Wei
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510000, China
| | - Ai Li
- Department of Clinical Medicine, The Second Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Li Wang
- Nephrology Department, Southern Medical University Affiliated Longhua People's Hospital, Shenzhen, China.
| | - Xiangqiu Chen
- Department of Urology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518100, China.
| |
Collapse
|
5
|
Chu DT, Ngo AD, Wu CC. Epigenetics in cancer development, diagnosis and therapy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 198:73-92. [PMID: 37225325 DOI: 10.1016/bs.pmbts.2023.01.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Cancer is a dangerous disease and one of the leading causes of death in the world. In 2020, there were nearly 10 million cancer deaths and approximately 20 million new cases. New cases and deaths from cancer are expected to increase further in the coming years. To have a deeper insight into the mechanism of carcinogenesis, epigenetics studies have been published and received much attention from scientists, doctors, and patients. Among alterations in epigenetics, DNA methylation and histone modification are studied by many scientists. They have been reported to be a major contributor in tumor formation and are involved in metastasis. From the understanding of DNA methylation and histone modification, effective, accurate and cost-effective methods for diagnosis and screening of cancer patients have been introduced. Furthermore, therapeutic approaches and drugs targeting altered epigenetics have also been clinically studied and have shown positive results in combating tumor progression. Several cancer drugs that rely on DNA methylation inactivation or histone modification have been approved by the FDA for the treatment of cancer patients. In summary, epigenetics changes such as DNA methylation or histone modification are take part in tumor growth, and they also have great prospect to study diagnostic and therapeutic methods of this dangerous disease.
Collapse
Affiliation(s)
- Dinh-Toi Chu
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam; Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam.
| | - Anh-Dao Ngo
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam
| | - Chia-Ching Wu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan; International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan; Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
6
|
Nandalur KR, Colvin R, Walker D, Nandalur SR, Seifman B, Gangwish D, Hafron J. Benign prostate hyperplasia as a potential protective factor against prostate cancer: Insights from a magnetic resonance imaging study of compositional characteristics. Prostate 2021; 81:1097-1104. [PMID: 34375453 DOI: 10.1002/pros.24207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 11/07/2022]
Abstract
PURPOSE The structural relationship between benign prostate hyperplasia (BPH) and prostate cancer (Pca) is controversial. The purpose of our study was to examine the association between quantitative prostate compositional metrics by magnetic resonance imaging (MRI) and Pca. METHODS We identified 405 patients who underwent prostate MRI and biopsy and/or prostatectomy from January 2019 to January 2021 at our institution. Segmentation volumetric methods were used to assess central gland (CG) and peripheral zone (PZ) volume. PZ mean thickness and mean apparent diffusion coefficient (ADC), marker of underlying histologic components, were measured. Multivariable logistic regression was performed with outcomes of ≥Grade Group (GG) 2 Pca and for multifocal disease. RESULTS On multivariable analysis, higher CG volumes were at lower odds of ≥GG2 disease (n = 227) (OR: 0.97, 95% CI 0.96-0.98, p < 0.0001), taking into account PZ volume (p = 0.18) and thickness (p = 0.70). For every one cc increase in CG volume, there was an approximately 3% decrease in odds of ≥GG2 disease. Similar findings were noted for multifocal disease (n = 180) (OR: 0.97, 95% CI 0.96-0.98, p < 0.0001). Notably, ADC of the normal PZ was not significantly associated with CG volume (p = 0.21) nor a predictor of disease (p = 0.49). CONCLUSIONS Increasing central gland volume, driven by BPH, is associated with lower odds of significant Pca, including multifocal disease, while PZ anatomic and histologic surrogate changes were noncontributory. Findings support BPH impediment of global tumor growth predicted by theoretical mechanobiological model. This potential stabilizing factor should be further studied for risk stratification and in consideration for BPH therapy.
Collapse
Affiliation(s)
- Kiran R Nandalur
- Department of Radiology and Molecular Imaging, Oakland University William Beaumont Hospital School of Medicine, Royal Oak, Michigan, USA
| | - Robert Colvin
- Department of Radiology and Molecular Imaging, Oakland University William Beaumont Hospital School of Medicine, Royal Oak, Michigan, USA
| | - David Walker
- Department of Radiology and Molecular Imaging, Oakland University William Beaumont Hospital School of Medicine, Royal Oak, Michigan, USA
| | - Sirisha R Nandalur
- Department of Radiation Oncology, Oakland University William Beaumont Hospital School of Medicine, Royal Oak, Michigan, USA
| | - Brian Seifman
- Department of Urology, Oakland University William Beaumont Hospital School of Medicine, Royal Oak, Michigan, USA
| | - David Gangwish
- Department of Urology, Oakland University William Beaumont Hospital School of Medicine, Royal Oak, Michigan, USA
| | - Jason Hafron
- Department of Urology, Oakland University William Beaumont Hospital School of Medicine, Royal Oak, Michigan, USA
| |
Collapse
|
7
|
Epigenetic reprogramming during prostate cancer progression: A perspective from development. Semin Cancer Biol 2021; 83:136-151. [PMID: 33545340 DOI: 10.1016/j.semcancer.2021.01.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/27/2021] [Accepted: 01/27/2021] [Indexed: 12/15/2022]
Abstract
Conrad Waddington's theory of epigenetic landscape epitomize the process of cell fate and cellular decision-making during development. Wherein the epigenetic code maintains patterns of gene expression in pluripotent and differentiated cellular states during embryonic development and differentiation. Over the years disruption or reprogramming of the epigenetic landscape has been extensively studied in the course of cancer progression. Cellular dedifferentiation being a key hallmark of cancer allow us to take cues from the biological processes involved during development. Here, we discuss the role of epigenetic landscape and its modifiers in cell-fate determination, differentiation and prostate cancer progression. Lately, the emergence of RNA-modifications has also furthered our understanding of epigenetics in cancer. The overview of the epigenetic code regulating androgen signalling, and progression to aggressive neuroendocrine stage of PCa reinforces its gene regulatory functions during the development of prostate gland as well as cancer progression. Additionally, we also highlight the clinical implications of cancer cell epigenome, and discuss the recent advancements in the therapeutic strategies targeting the advanced stage disease.
Collapse
|
8
|
Lin ZM, Fan DD, Jin S, Liu ZL, Niu YN. Methylated CpG dinucleotides in the 5-α reductase 2 gene may explain finasteride resistance in benign prostatic enlargement patients. Asian J Androl 2021; 23:266-272. [PMID: 33243958 PMCID: PMC8152415 DOI: 10.4103/aja.aja_63_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The inhibition of 5-α reductase type 2 (SRD5A2) by finasteride is commonly used for the management of urinary obstruction resulting from benign prostatic enlargement (BPE). Certain BPE patients showing no SRD5A2 protein expression are resistant to finasteride therapy. Our previous work showed that methylated cytosine-phosphate-guanine (CpG) islands in the SRD5A2 gene might account for the absence or reduction of SRD5A2 protein expression. Here, we found that the expression of the SRD5A2 protein was variable and that weak expression of the SRD5A2 protein (scored 0–100) occurred in 10.0% (4/40) of benign adult prostates. We showed that the expression of SRD5A2 was negatively correlated with DNA methyltransferase 1 (DNMT1) expression. In vitro SRD5A2-negative BPH-1 cells were resistant to finasteride treatment, and SRD5A2 was re-expressed in BPH-1 cells when SRD5A2 was demethylated by 5-Aza-2'-deoxycytidine (5-Aza-CdR) or N-phthalyl-L-tryptophan (RG108). Furthermore, we determined the exact methylation ratios of CpG dinucleotides in a CpG island of SRD5A2 through MassArray quantitative methylation analysis. Ten methylated CpG dinucleotides, including four CpG dinucleotides in the promoter and six CpG dinucleotides in the first exon, were found in a CpG island located from −400 bp to +600 bp in SRD5A2, which might lead to the silencing of SRD5A2 and the absence or reduction of SRD5A2 protein expression. Finasteride cannot exert a therapeutic effect on patients lacking SRD5A2, which may partially account for the resistance to finasteride observed in certain BPE patients.
Collapse
Affiliation(s)
- Zhe-Min Lin
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100016, China
| | - Dong-Dong Fan
- Department of Urology, Beijing Miyun Teaching Hospital, Capital Medical University, Beijing 101500, China
| | - Song Jin
- Department of Urology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Zhan-Liang Liu
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100016, China
| | - Yi-Nong Niu
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100016, China
| |
Collapse
|
9
|
Olayinka ET, Adewole KE. In vivo and in silico evaluation of the ameliorative effect of hesperidin on finasteride-induced testicular oxidative stress in Wistar rats. Toxicol Mech Methods 2020; 31:81-89. [PMID: 33003968 DOI: 10.1080/15376516.2020.1831123] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Finasteride used for treating benign prostatic hyperplasia is associated with undesirable side effects via oxidative stress related mechanisms. This study employed in vivo and in silico methods to investigate the protective role of hesperidin against testicular toxicity induced by finasteride and the possible molecular mechanisms involved. Male Wistar rats were randomized into four groups of six animals each. Group I (control) were administered distilled water, group II received finasteride (3.1 mg/kg bw), group III received hesperidin (100 mg/kg bw), while group IV were co-administered finasteride and hesperidin. Administration was by gavage for 14 days. The binding propensities of finasteride and hesperidin for 5α-reductase were assessed using in silico docking approach. Finasteride administration caused significant reductions of sperm motility, volume, count, and live/dead ratio, with significant increase in numbers of abnormal sperms. Finasteride treatment also resulted in diminished activities of superoxide dismutase, catalase and glutathione-S-transferase, significant reduction in the concentration of reduced glutathione and ascorbic acid, and increased testicular malondialdehyde level relative to control. Moreover, significant increase in the activities of testicular lactate dehydrogenase and γ-glutamyl transferase was observed, with significant decrease in the activities of acid phosphatase and alkaline phosphatase relative to finasteride-treated rats. Furthermore, hesperidin exhibited favorable binding affinity for 5α -reductase (5AR) in silico compared to finasteride. Co-administration with hesperidin ameliorated finasteride-induced testicular damage by suppressing oxidative stress indices, enhancing antioxidant status, improving sperm parameters and alterations in the activities of marker enzymes, as well as possibly inhibiting the binding of finasteride to 5AR.
Collapse
Affiliation(s)
- Ebenezer Tunde Olayinka
- Biochemistry Unit, Department of Chemical Sciences, Ajayi Crowther University, PMB, Oyo, Oyo State, Nigeria
| | - Kayode Ezekiel Adewole
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Medical Sciences, Ondo, Ondo State, Nigeria
| |
Collapse
|
10
|
Mehdi A, Cheishvili D, Arakelian A, Bismar TA, Szyf M, Rabbani SA. DNA methylation signatures of Prostate Cancer in peripheral T-cells. BMC Cancer 2020; 20:588. [PMID: 32576165 PMCID: PMC7310561 DOI: 10.1186/s12885-020-07078-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 06/15/2020] [Indexed: 01/03/2023] Open
Abstract
Background Prostate Cancer (PCa) is the second most common cancer in men where advancements have been made for early detection using imaging techniques, however these are limited by lesion size. Immune surveillance has emerged as an effective approach for early detection and to monitor disease progression. In recent studies, we have shown that host peripheral blood immune cells undergo changes in DNA methylation in liver and breast cancer. Methods In the current study, we examined the DNA methylation status of peripheral blood T cells of men with positive biopsy for PCa versus men with negative biopsy having benign prostate tissue, defined as controls. T cells DNA was isolated and subjected to Illumina Infinium methylation EPIC array and validated using Illumina amplicon sequencing and pyrosequencing platforms. Results Differential methylation of 449 CG sites between control and PCa T cell DNA showed a correlation with Gleason score (p < 0.05). Two hundred twenty-three differentially methylated CGs between control and PCa (∆ß +/− 10%, p < 0.05), were enriched in pathways involved in immune surveillance system. Three CGs which were found differentially methylated following DMP (Differentially methylated probes) analysis of ChAMP remained significant after BH (Benjamini-Hochberg) correction, of which, 2 CGs were validated. Predictive ability of combination of these 3 CGs (polygenic methylation score, PMS) to detect PCa had high sensitivity, specificity and overall accuracy. PMS also showed strong positive correlation with Gleason score and tumor volume of PCa patients. Conclusions Results from the current study provide for the first-time a potential role of DNA methylation changes in peripheral T cells in PCa. This non-invasive methodology may allow for early intervention and stratification of patients into different prognostic groups to reduce PCa associated morbidity from repeat invasive prostate biopsies and design therapeutic strategy to reduce PCa associated mortality.
Collapse
Affiliation(s)
- Ali Mehdi
- Department of Medicine, McGill University, Montreal, Quebec, Canada.,Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - David Cheishvili
- HKG Epitherapeutics, Hong Kong, China.,Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - Ani Arakelian
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Tarek A Bismar
- Departments of Pathology & Laboratory Medicine, Oncology, Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Moshe Szyf
- Department of Pharmacology, McGill University, Montreal, Quebec, Canada
| | - Shafaat A Rabbani
- Department of Medicine, McGill University, Montreal, Quebec, Canada. .,Department of Human Genetics, McGill University, Montreal, Quebec, Canada. .,McGill University Health Centre, 1001 Décarie Blvd. (Glen site), Room EM1.3232, Montréal, QC, H4A3J1, Canada.
| |
Collapse
|
11
|
Obesity-associated inflammation induces androgenic to estrogenic switch in the prostate gland. Prostate Cancer Prostatic Dis 2020; 23:465-474. [PMID: 32029929 DOI: 10.1038/s41391-020-0208-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 01/14/2020] [Accepted: 01/28/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND OBJECTIVE Our patient cohort revealed that obesity is strongly associated with steroid-5α reductase type 2 (SRD5A2) promoter methylation and reduced protein expression. The underlying mechanism of prostatic growth in this population is poorly understood. Here we addressed the question of how obesity, inflammation, and steroid hormones affect the development of benign prostatic hyperplasia (BPH). MATERIAL AND METHODS We used preadipocytes, macrophages, primary human prostatic stromal cells, prostate tissues from high-fat diet-induced obese mice, and 35 prostate specimens that were collected from patients who underwent transurethral resection of the prostate (TURP). RNA was isolated and quantified with RT-PCR. Genome DNA was extracted and SRD5A2 promoter methylation was determined. Sex hormones were determined by high-performance liquid chromatography-tandem mass spectrometry. Protein was extracted and determined by ELISA test. RESULTS In prostatic tissues with obesity, the levels of inflammatory mediators were elevated. SRD5A2 promoter methylation was promoted, but SRD5A2 expression was inhibited. Inflammatory mediators and saturated fatty acid synergistically regulated aromatase activity. Obesity promoted an androgenic to estrogenic switch in the prostate. CONCLUSIONS Our findings suggest that obesity-associated inflammation induces androgenic to estrogenic switch in the prostate gland, which may serve as an effective strategy for alternative therapies for management of lower urinary tract symptoms associated with BPH in select individuals.
Collapse
|
12
|
Strmiska V, Michalek P, Lackova Z, Guran R, Krizkova S, Vanickova L, Zitka O, Stiborova M, Eckschlager T, Klejdus B, Pacik D, Tvrdikova E, Keil C, Haase H, Adam V, Heger Z. Sarcosine is a prostate epigenetic modifier that elicits aberrant methylation patterns through the SAMe-Dnmts axis. Mol Oncol 2019; 13:1002-1017. [PMID: 30628163 PMCID: PMC6487735 DOI: 10.1002/1878-0261.12439] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/23/2018] [Accepted: 12/30/2018] [Indexed: 12/31/2022] Open
Abstract
DNA hypermethylation is one of the most common epigenetic modifications in prostate cancer (PCa). Several studies have delineated sarcosine as a PCa oncometabolite that increases the migration of malignant prostate cells while decreasing their doubling time. Here, we show that incubation of prostate cells with sarcosine elicited the upregulation of sarcosine N‐demethylation enzymes, sarcosine dehydrogenase and pipecolic acid oxidase. This process was accompanied by a considerable increase in the production of the major methyl‐donor S‐adenosylmethionine (SAMe), together with an elevation of cellular methylation potential. Global DNA methylation analyses revealed increases in methylated CpG islands in distinct prostate cell lines incubated with sarcosine, but not in cells of nonprostate origin. This phenomenon was further associated with marked upregulation of DNA methyltransferases (Dnmts). Epigenetic changes were recapitulated through blunting of Dnmts using the hypomethylating agent 5‐azacytidine, which was able to inhibit sarcosine‐induced migration of prostate cells. Moreover, spatial mapping revealed concomitant increases in sarcosine, SAMe and Dnmt1 in histologically confirmed malignant prostate tissue, but not in adjacent or nonmalignant tissue, which is in line with the obtained in vitro data. In summary, we show here for the first time that sarcosine acts as an epigenetic modifier of prostate cells and that this may contribute to its oncometabolic role.
Collapse
Affiliation(s)
- Vladislav Strmiska
- Department of Chemistry and Biochemistry, Mendel University in Brno, Czech Republic
| | - Petr Michalek
- Department of Chemistry and Biochemistry, Mendel University in Brno, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Czech Republic
| | - Zuzana Lackova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Czech Republic
| | - Roman Guran
- Department of Chemistry and Biochemistry, Mendel University in Brno, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Czech Republic
| | - Sona Krizkova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Czech Republic
| | - Lucie Vanickova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Czech Republic
| | - Ondrej Zitka
- Department of Chemistry and Biochemistry, Mendel University in Brno, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Czech Republic
| | - Marie Stiborova
- Department of Biochemistry, Faculty of Science, Charles University, Prague 2, Czech Republic
| | - Tomas Eckschlager
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University, University Hospital Motol, Prague 5, Czech Republic
| | - Borivoj Klejdus
- Department of Chemistry and Biochemistry, Mendel University in Brno, Czech Republic.,Central European Institute of Technology, Mendel University in Brno, Czech Republic
| | - Dalibor Pacik
- Department of Urology, University Hospital Brno, Brno, Czech Republic.,Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Eliska Tvrdikova
- Department of Pathology, University Hospital Brno, Czech Republic
| | - Claudia Keil
- Department of Food Chemistry and Toxicology, Technical University of Berlin, Germany
| | - Hajo Haase
- Department of Food Chemistry and Toxicology, Technical University of Berlin, Germany
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Czech Republic
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Czech Republic
| |
Collapse
|
13
|
5hmC Level Predicts Biochemical Failure Following Radical Prostatectomy in Prostate Cancer Patients with ERG Negative Tumors. Int J Mol Sci 2019; 20:ijms20051025. [PMID: 30818754 PMCID: PMC6429366 DOI: 10.3390/ijms20051025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 12/19/2022] Open
Abstract
This study aimed to validate whether 5-hydroxymethylcytosine (5hmC) level in combination with ERG expression is a predictive biomarker for biochemical failure (BF) in men undergoing radical prostatectomy (RP) for prostate cancer (PCa). The study included 592 PCa patients from two consecutive Danish RP cohorts. 5hmC level and ERG expression were analyzed using immunohistochemistry in RP specimens. 5hmC was scored as low or high and ERG was scored as negative or positive. Risk of BF was analyzed using stratified cumulative incidences and multiple cause-specific Cox regression using competing risk assessment. Median follow-up was 10 years (95% CI: 9.5–10.2). In total, 246 patients (41.6%) had low and 346 patients (58.4%) had high 5hmC level. No significant association was found between 5hmC level or ERG expression and time to BF (p = 0.2 and p = 1.0, respectively). However, for men with ERG negative tumors, high 5hmC level was associated with increased risk of BF following RP (p = 0.01). In multiple cause-specific Cox regression analyses of ERG negative patients, high 5hmC expression was associated with time to BF (HR: 1.8; 95% CI: 1.2–2.7; p = 0.003). In conclusion, high 5hmC level was correlated with time to BF in men with ERG negative PCa, which is in accordance with previous results.
Collapse
|
14
|
Zhang Y, Yan L, Yao W, Chen K, Xu H, Ye Z. Integrated Analysis of Genetic Abnormalities of the Histone Lysine Methyltransferases in Prostate Cancer. Med Sci Monit 2019; 25:193-239. [PMID: 30616239 PMCID: PMC6330996 DOI: 10.12659/msm.912294] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background The histone methyltransferase (HMT) family includes histone lysine methyltransferases (HKMTs) and histone/protein arginine methyltransferases (PRMTs). The role of HMT gene variants in prostate cancer remains unknown. Therefore, this study aimed to evaluate HMT gene variants in the pathogenesis and prognosis of human prostate cancer, using in vitro cell studies and bioinformatics analysis. Material/Methods Integrative bioinformatics analysis of the expression of 51 HMT genes in human prostate cancer was based on datasets from the Cancer Genome Atlas (TCGA). Correlation and regression analysis were used to identify critical HMTs in prostate cancer. Kaplan-Meier and the area under the receiver operating characteristics curve (AUROC) were performed to evaluate the function of the HMTs on prognosis. Gene expression and function of 22Rv1 human prostate carcinoma cells were studied. Results The HMT genes identified to have a role in the pathogenesis of prostate cancer included the EZH2, SETD5, PRDM12, NSD1, SETD6, SMYD1, and the WHSC1L1 gene. The EZH2, SETD5, and SMYD1 genes were selected as a prognostic panel, with the SUV420H2 HMT gene. SETD2, NSD1, and ASH1L were identified as critical genes in the development of castration-resistant prostate cancer (CRPC), similar to mixed-lineage leukemia (MLL) complex family members. Knockdown of the SETD5 gene in 22Rv1 prostate carcinoma cells in vitro inhibited cancer cell growth and migration. Conclusions HMT gene variants may have a role in the pathogenesis of prostate cancer. Future studies may determine the role of HMT genes as prognostic biomarkers in patients with prostate cancer.
Collapse
Affiliation(s)
- Yangjun Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, AL, China (mainland).,Institute of Urology of Hubei Province, Wuhan, Hubei, China (mainland)
| | - Libin Yan
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland).,Institute of Urology of Hubei Province, Wuhan, Hubei, China (mainland)
| | - Weimin Yao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, AL, China (mainland).,Institute of Urology of Hubei Province, Wuhan, Hubei, China (mainland)
| | - Ke Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, AL, China (mainland).,Institute of Urology of Hubei Province, Wuhan, Hubei, China (mainland)
| | - Hua Xu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, AL, China (mainland).,Institute of Urology of Hubei Province, Wuhan, Hubei, China (mainland)
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, AL, China (mainland).,Institute of Urology of Hubei Province, Wuhan, Hubei, China (mainland)
| |
Collapse
|
15
|
Lian XY, Ma JC, Zhou JD, Zhang TJ, Wu DH, Deng ZQ, Zhang ZH, Li XX, He PF, Yan Y, Lin J, Qian J. Hypermethylation of ITGBL1 is associated with poor prognosis in acute myeloid leukemia. J Cell Physiol 2018; 234:9438-9446. [PMID: 30317626 DOI: 10.1002/jcp.27629] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/24/2018] [Indexed: 12/22/2022]
Abstract
The current study was aimed to investigate integrin beta-like 1 (ITGBL1) methylation pattern and its clinical relevance in patients with acute myeloid leukemia (AML). Real-time methylation-specific polymerase chain reaction (PCR; RQ-MSP) and bisulfite sequencing PCR (BSP) were performed to detect the methylation of ITGBL1 promoter. Real-time quantitative PCR (RT-qPCR) was performed to analyze ITGBL1 transcript level. The results showed that ITGBL1 methylation level in 131 patients with AML was significantly higher than 29 controls (p < 0.001). The ITGBL1-hypermethylated group tended to have a higher bone marrow (BM) blasts ( p = 0.076). Meanwhile, ITGBL1-hypermethylated patients tended to have a lower complete remission (CR) rate ( p = 0.102). ITGBL1-hypermethylated patients had significantly shorter overall survival (OS) and leukemia-free survival (LFS) than ITGBL1 hypomethylated patients in whole AML cohort ( p = 0.009 and 0.043, respectively) and patients with nonacute promyelocytic leukemia (APL ; p = 0.023 and 0.039, respectively). Multivariate analysis confirmed that the ITGBL1 methylation served as an independent prognostic factor in both patients with whole-cohort AML ( p = 0.030) and patients with non-APL ( p = 0.020). Furthermore, the ITGBL1 methylation level was significantly decreased in follow-up AML patients who achieved complete remission after induction therapy ( P = 0.001). ITGBL1 methylation negatively correlated with ITGBL1 expression in patients with AML ( R = -0.328, p = 0.008). Moreover, demethylation of ITGBL1 could increase the ITGBL1 expression in the K562 leukemic cell line ( p < 0.05). In conclusion, the ITGBL1 hypermethylation is a potential biomarker for predicting prognosis and monitoring disease status in patients with AML.
Collapse
Affiliation(s)
- Xin-Yue Lian
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.,Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ji-Chun Ma
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, China
| | - Jing-Dong Zhou
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, China
| | - Ting-Juan Zhang
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, China
| | - De-Hong Wu
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, China.,Department of Hematology, The Third People's Hospital of Kunshan City, Kunshan, Jiangsu, China
| | - Zhao-Qun Deng
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, China
| | - Zhi-Hui Zhang
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, China
| | - Xi-Xi Li
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, China
| | - Pin-Fang He
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, China
| | - Yang Yan
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, China.,School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jiang Lin
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, China
| | - Jun Qian
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, China
| |
Collapse
|
16
|
Lian XY, Zhang W, Wu DH, Ma JC, Zhou JD, Zhang ZH, Wen XM, Xu ZJ, Lin J, Qian J. Methylation-independent ITGA2 overexpression is associated with poor prognosis in de novo acute myeloid leukemia. J Cell Physiol 2018; 233:9584-9593. [PMID: 30132837 DOI: 10.1002/jcp.26866] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 05/22/2018] [Indexed: 12/13/2022]
Abstract
Previous studies have been indicated that integrin α2 (ITGA2) may be important in cell migration, invasion, survival, and angiogenesis. However, the correlation between ITGA2 expression and acute myeloid leukemia (AML) is still unclear. Real-time quantitative polymerase chain reaction was carried out to analyze ITGA2 messenger RNA level. Methylation-specific polymerase chain reaction (PCR) and bisulfite sequencing PCR were performed to detect the methylation of ITGA2 promoter. ITGA2 expression was significantly upregulated in 134 de novo AML patients compared with 33 controls (p = 0.007). ITGA2high group had markedly lower complete remission (CR) rate than ITGA2low group (p = 0.011). Furthermore, the overall survival in ITGA2high patients was significantly shorter than ITGA2low patients throughout AML cohort, non-acute promyelocytic leukemia (APL) and cytogenetic normal-AML (p = 0.001, 0.002, and 0.044, respectively). Multivariate analysis confirmed that ITGA2 overexpression served as an independent prognostic factor in both whole-cohort AML patients (p = 0.018) and non-APL AML patients (p = 0.021). Besides, ITGA2 expression level was significantly decreased in AML patients after CR (p = 0.011), and was returned at the time of relapse phase (p = 0.021). Moreover, unmethylated ITGA2 promoter was identified in normal controls, leukemia cell lines, and primary leukemia cells with low or high ITGA2 expression. In conclusions, methylation-independent ITGA2 overexpression is associated with poor prognosis in AML.
Collapse
Affiliation(s)
- Xin-Yue Lian
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, China
| | - Wei Zhang
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, China
| | - De-Hong Wu
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, China.,Department of Hematology, The Third People's Hospital of Kunshan City, Kunshan, China
| | - Ji-Chun Ma
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, China.,Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Jing-Dong Zhou
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, China
| | - Zhi-Hui Zhang
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, China
| | - Xiang-Mei Wen
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, China.,Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Zi-Jun Xu
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, China.,Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Jiang Lin
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, China.,Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Jun Qian
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, China
| |
Collapse
|
17
|
Kang PM, Kim YJ, Seo WT, Kang SH, Kim TS, Chun BK, Seo WI, Jeong JY, Chung JI. Correlation between 5-α reductase type 2 protein expression and methylation of 5-α reductase type 2 promotor gene of benign prostatic hyperplasia. World J Urol 2018; 37:709-718. [PMID: 30069579 DOI: 10.1007/s00345-018-2422-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 07/25/2018] [Indexed: 12/14/2022] Open
Abstract
PURPOSE The enzyme 5-α reductase type 2 (5-AR 2) plays a key role in the development and maintenance of the prostate gland. We evaluated the level 5-AR 2 protein expression and the relationship between methylation of the 5-AR 2 gene-promoter and 5-AR 2 protein expression of benign prostatic hyperplasia (BPH). MATERIALS AND METHODS A total of 37 prostate samples were evaluated. These included 22 samples from men undergoing transurethral prostate resections and 15 non-cancerous transition-zone human prostate tissue samples taken following radical prostatectomy. We quantified 5-AR 2 protein expression and gene-promoter methylation status using common assay procedures. Clinical variables included age, body mass index (BMI), prostate-specific antigen (PSA) levels, lipid profiles, and prostate volumes. Univariate and multivariate statistical analyses were performed followed by stepwise logistic regression modeling. RESULTS We were able to extract DNA from 36 of the 37 tissue samples and 10 of these (28%) did not express the 5-AR 2 protein. In total, 26 patients (72%) had methylated 5-AR 2 promoter-regions. There was a strong correlation between methylation of the 5-AR 2 promoter-regions and low-absent 5-AR 2 protein expression (p = 0.0003). Increasing age significantly predicted methylation status and protein expression level (p = 0.013). CONCLUSIONS The level of 5-AR 2 protein expression varies among prostate tissue samples. Methylation of the 5-AR 2 gene-promoter may account for low or absent expression of 5-AR 2 in adult human prostate tissues. Increased age correlates with increased 5-AR 2 gene-promoter methylation and decreased protein expression in men with BPH.
Collapse
Affiliation(s)
- Pil Moon Kang
- Department of Urology, Kosin Gospel Hospital, Kosin University College of Medicine, Busan, Korea
| | - Young Jin Kim
- Department of Biochemistry, Kosin University College of Medicine, 262 Gamcheon-ro, Seo-gu, Busan, 49267, Korea.,Cancer Research Institute, Kosin University College of Medicine, Busan, Korea
| | - Won Tae Seo
- Department of Urology, Kosin Gospel Hospital, Kosin University College of Medicine, Busan, Korea
| | - Su Hwan Kang
- Department of Urology, Kosin Gospel Hospital, Kosin University College of Medicine, Busan, Korea
| | - Taek Sang Kim
- Department of Urology, Kosin Gospel Hospital, Kosin University College of Medicine, Busan, Korea
| | - Bong Kwon Chun
- Department of Pathology, Kosin University College of Medicine, Busan, Korea
| | - Won Ik Seo
- Department of Urology, Busan Paik Hospital, Inje University College of Medicine, Gaegeum-dong, Busanjin-gu, Busan, 614-735, Korea
| | - Jee-Yeong Jeong
- Department of Biochemistry, Kosin University College of Medicine, 262 Gamcheon-ro, Seo-gu, Busan, 49267, Korea. .,Cancer Research Institute, Kosin University College of Medicine, Busan, Korea.
| | - Jae Il Chung
- Department of Urology, Busan Paik Hospital, Inje University College of Medicine, Gaegeum-dong, Busanjin-gu, Busan, 614-735, Korea.
| |
Collapse
|
18
|
Damodaran S, Damaschke N, Gawdzik J, Yang B, Shi C, Allen GO, Huang W, Denu J, Jarrard D. Dysregulation of Sirtuin 2 (SIRT2) and histone H3K18 acetylation pathways associates with adverse prostate cancer outcomes. BMC Cancer 2017; 17:874. [PMID: 29262808 PMCID: PMC5738829 DOI: 10.1186/s12885-017-3853-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 11/28/2017] [Indexed: 12/15/2022] Open
Abstract
Background Histones undergo extensive post-translational modifications and this epigenetic regulation plays an important role in modulating transcriptional programs capable of driving cancer progression. Acetylation of histone H3K18, associated with gene activation, is enhanced by P300 and opposed by the deacetylase Sirtuin2 (SIRT2). As these enzymes represent an important target for cancer therapy, we sought to determine whether the underlying genes are altered during prostate cancer (PCa) progression. Methods Tissue microarrays generated from 71 radical prostatectomy patients were initially immunostained for H3K18Ac, P300 and SIRT2. Protein levels were quantified using VECTRA automation and correlated with clinicopathologic parameters. The Cancer Genome Atlas (TGCA, n = 499) and Gene Expression Omnibus (n = 504) databases were queried for expression, genomic and clinical data. Statistics were performed using SPSSv23. Results Nuclear histone H3K18Ac staining increases in primary cancer (p = 0.05) and further in metastases (p < 0.01) compared to benign on tissue arrays. P300 protein expression increases in cancer (p = 0.04) and metastases (p < 0.001). A progressive decrease in nuclear SIRT2 staining occurs comparing benign to cancer or metastases (p = 0.04 and p = 0.03 respectively). Decreased SIRT2 correlates with higher grade cancer (p = 0.02). Time to Prostate Specific Antigen (PSA) recurrence is shorter in patients exhibiting high compared to low H3K18Ac expression (350 vs. 1542 days respectively, P = 0.03). In GEO, SIRT2 mRNA levels are lower in primary and metastatic tumors (p = 0.01 and 0.001, respectively). TGCA analysis demonstrates SIRT2 deletion in 6% and increasing clinical stage, positive margins and lower PSA recurrence-free survival in patients with SIRT2 loss/deletion (p = 0.01, 0.04 and 0.04 respectively). In this dataset, a correlation between decreasing SIRT2 and increasing P300 mRNA expression occurs in tumor samples (R = −0.46). Conclusions In multiple datasets, decreases in SIRT2 expression portend worse clinicopathologic outcomes. Alterations in SIRT2-H3K18Ac suggest altered P300 activity and identify a subset of tumors that could benefit from histone deacetylation inhibition. Electronic supplementary material The online version of this article (10.1186/s12885-017-3853-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shivashankar Damodaran
- Department of Urology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53705, USA
| | - Nathan Damaschke
- Department of Urology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53705, USA
| | - Joseph Gawdzik
- Department of Urology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53705, USA
| | - Bing Yang
- Department of Urology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53705, USA
| | - Cedric Shi
- Department of Urology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53705, USA
| | - Glenn O Allen
- Department of Urology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53705, USA
| | - Wei Huang
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - John Denu
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI, 53705, USA.,Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI, 53706, USA.,Wisconsin Institute for Discovery and the Morgridge Institute for Research, University of Wisconsin, Madison, WI, 53715, USA
| | - David Jarrard
- Department of Urology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53705, USA. .,Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI, 53705, USA. .,Molecular and Environmental Toxicology Program, University of Wisconsin, Madison, WI, 53706, USA. .,John P. Livesey Chair in Urologic Oncology, Associate Director Translational Research, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, 7037 WIMR, 1111, Highland, Avenue Madison WI, 53705, USA.
| |
Collapse
|
19
|
Falco M, Palma G, Rea D, De Biase D, Scala S, D'Aiuto M, Facchini G, Perdonà S, Barbieri A, Arra C. Tumour biomarkers: homeostasis as a novel prognostic indicator. Open Biol 2016; 6:160254. [PMID: 27927793 PMCID: PMC5204124 DOI: 10.1098/rsob.160254] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/10/2016] [Indexed: 12/15/2022] Open
Abstract
The term 'personalized medicine' refers to a medical procedure that consists in the grouping of patients based on their predicted individual response to therapy or risk of disease. In oncologic patients, a 'tailored' therapeutic approach may potentially improve their survival and well-being by not only reducing the tumour, but also enhancing therapeutic response and minimizing the adverse effects. Diagnostic tests are often used to select appropriate and optimal therapies that rely both on patient genome and other molecular/cellular analysis. Several studies have shown that lifestyle and environmental factors can influence the epigenome and that epigenetic events may be involved in carcinogenesis. Thus, in addition to traditional biomarkers, epigenetic factors are raising considerable interest, because they could potentially be used as an excellent tool for cancer diagnosis and prognosis. In this review, we summarize the role of conventional cancer genetic biomarkers and their association with epigenomics. Furthermore, we will focus on the so-called 'homeostatic biomarkers' that result from the physiological response to cancer, emphasizing the concept that an altered 'new' homeostasis influence not only tumour environment, but also the whole organism.
Collapse
Affiliation(s)
- Michela Falco
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Giuseppe Palma
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Domenica Rea
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Davide De Biase
- Department of Veterinary Medicine and Animal Production, University of Naples 'Federico II', Via Delpino 1, 80137 Naples, Italy
| | - Stefania Scala
- Molecular lmmunology and Immuneregulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, IRCCS Naples 'Fondazione G. Pascale', Naples, italy, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Massimiliano D'Aiuto
- Division of Breast Surgery, Department of Breast Disease, National Cancer Institute, IRCCS, 'Fondazione Pascale', Naples, Italy
| | - Gaetano Facchini
- Division of Medical Oncology, Department of Uro-Gynaecological Oncology, , Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione G. Pascale', IRCCS, 80131 Naples, Italy
| | - Sisto Perdonà
- Department of Urology, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione G. Pascale', IRCCS, 80131 Naples, Italy
| | - Antonio Barbieri
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Claudio Arra
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| |
Collapse
|
20
|
Scholpa NE, Kolli RT, Moore M, Arnold RD, Glenn TC, Cummings BS. Nephrotoxicity of epigenetic inhibitors used for the treatment of cancer. Chem Biol Interact 2016; 258:21-9. [PMID: 27543423 PMCID: PMC5045804 DOI: 10.1016/j.cbi.2016.08.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/21/2016] [Accepted: 08/15/2016] [Indexed: 12/25/2022]
Abstract
This study determined the anti-neoplastic activity and nephrotoxicity of epigenetic inhibitors in vitro. The therapeutic efficacy of epigenetic inhibitors was determined in human prostate cancer cells (PC-3 and LNCaP) using the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine (5-Aza) and the histone deacetylase inhibitor trichostatin A (TSA). Cells were also treated with carbamazepine (CBZ), an anti-convulsant with histone deacetylase inhibitor-like properties. 5-Aza, TSA or CBZ alone did not decrease MTT staining in PC-3 or LNCaP cells after 48 h. In contrast, docetaxel, a frontline chemotherapeutic induced concentration-dependent decreases in MTT staining. Pretreatment with 5-Aza or TSA increased docetaxel-induced cytotoxicity in LNCaP cells, but not PC-3 cells. TSA pretreatment also increased cisplatin-induced toxicity in LNCaP cells. Carfilzomib (CFZ), a protease inhibitor approved for the treatment of multiple myeloma had minimal effect on LNCaP cell viability, but reduced MTT staining 50% in PC-3 cells compared to control, and pretreatment with 5-Aza further enhanced toxicity. Treatment of normal rat kidney (NRK) and human embryonic kidney 293 (HEK293) cells with the same concentrations of epigenetic inhibitors used in prostate cancer cells significantly decreased MTT staining in all cell lines after 48 h. Interestingly, we found that the toxicity of epigenetic inhibitors to kidney cells was dependent on both the compound and the stage of cell growth. The effect of 5-Aza and TSA on DNA methyltransferase and histone deacetylase activity, respectively, was confirmed by assessing the methylation and acetylation of the CDK inhibitor p21. Collectively, these data show that combinatorial treatment with epigenetic inhibitors alters the efficacy of chemotherapeutics in cancer cells in a compound- and cell-specific manner; however, this treatment also has the potential to induce nephrotoxic cell injury.
Collapse
Affiliation(s)
- N E Scholpa
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, USA
| | - R T Kolli
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, USA
| | - M Moore
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, USA
| | - R D Arnold
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - T C Glenn
- Department of Environmental Health Science, University of Georgia, Athens, GA 30602, USA
| | - B S Cummings
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
21
|
Amacher DE. A 2015 survey of established or potential epigenetic biomarkers for the accurate detection of human cancers. Biomarkers 2016; 21:387-403. [PMID: 26983778 DOI: 10.3109/1354750x.2016.1153724] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Context The silencing or activation of cancer-associated genes by epigenetic mechanisms can ultimately lead to the clonal expansion of cancer cells. Objective The aim of this review is to summarize all relevant epigenetic biomarkers that have been proposed to date for the diagnosis of some prevalent human cancers. Methods A Medline search for the terms epigenetic biomarkers, human cancers, DNA methylation, histone modifications and microRNAs was performed. Results One hundred fifty-seven relevant publications were found and reviewed. Conclusion To date, a significant number of potential epigenetic cancer biomarkers of human cancer have been investigated, and some have advanced to clinical implementation.
Collapse
|
22
|
Verma M. The Role of Epigenomics in the Study of Cancer Biomarkers and in the Development of Diagnostic Tools. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 867:59-80. [PMID: 26530360 DOI: 10.1007/978-94-017-7215-0_5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Epigenetics plays a key role in cancer development. Genetics alone cannot explain sporadic cancer and cancer development in individuals with no family history or a weak family history of cancer. Epigenetics provides a mechanism to explain the development of cancer in such situations. Alterations in epigenetic profiling may provide important insights into the etiology and natural history of cancer. Because several epigenetic changes occur before histopathological changes, they can serve as biomarkers for cancer diagnosis and risk assessment. Many cancers may remain asymptomatic until relatively late stages; in managing the disease, efforts should be focused on early detection, accurate prediction of disease progression, and frequent monitoring. This chapter describes epigenetic biomarkers as they are expressed during cancer development and their potential use in cancer diagnosis and prognosis. Based on epigenomic information, biomarkers have been identified that may serve as diagnostic tools; some such biomarkers also may be useful in identifying individuals who will respond to therapy and survive longer. The importance of analytical and clinical validation of biomarkers is discussed, along with challenges and opportunities in this field.
Collapse
Affiliation(s)
- Mukesh Verma
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute (NCI), National Institutes of Health (NIH), Suite# 4E102. 9609 Medical Center Drive, MSC 9763, Bethesda, MD, 20892-9726, USA.
| |
Collapse
|
23
|
Strand SH, Hoyer S, Lynnerup AS, Haldrup C, Storebjerg TM, Borre M, Orntoft TF, Sorensen KD. High levels of 5-hydroxymethylcytosine (5hmC) is an adverse predictor of biochemical recurrence after prostatectomy in ERG-negative prostate cancer. Clin Epigenetics 2015; 7:111. [PMID: 26478752 PMCID: PMC4608326 DOI: 10.1186/s13148-015-0146-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 10/02/2015] [Indexed: 12/14/2022] Open
Abstract
Background Prostate cancer (PC) can be stratified into distinct molecular subtypes based on TMPRSS2-ERG gene fusion status, but its potential prognostic value remains controversial. Likewise, routine clinicopathological features cannot clearly distinguish aggressive from indolent tumors at the time of diagnosis; thus, new prognostic biomarkers are urgently needed. The DNA methylation variant 5-hydroxymethylcytosine (5hmC, an oxidized derivative of 5-methylcytosine) has recently emerged as a new diagnostic and/or prognostic biomarker candidate for several human malignancies. However, this remains to be systematically investigated for PC. In this study, we determined 5hmC levels in 311 PC (stratified by ERG status) and 228 adjacent non-malignant (NM) prostate tissue specimens by immunohistochemical analysis of a tissue microarray, representing a large radical prostatectomy (RP) cohort with long clinical follow-up. We investigated possible correlations between 5hmC and routine clinicopathological variables and assessed the prognostic potential of 5hmC by Kaplan-Meier and uni- and multivariate Cox regression analyses in ERG+ (n = 178) vs. ERG− (n = 133) PCs using biochemical recurrence (BCR) as endpoint. Results We observed a borderline significant (p = 0.06) reduction in 5hmC levels in PC compared to NM tissue samples, which was explained by a highly significant (p < 0.001) loss of 5hmC in ERG− PCs. ERG status was not predictive of BCR in this cohort (p = 0.73), and no significant association was found between BCR and 5hmC levels in ERG+ PCs (p = 0.98). In contrast, high 5hmC immunoreactivity was a significant adverse predictor of BCR after RP in ERG− PCs, independent of Gleason score, pathological tumor stage, surgical margin status, and pre-operative prostate-specific antigen (PSA) level (hazard ratio (HR) (95 % confidence interval (CI)): 1.62 (1.15–2.28), p = 0.006). Conclusions This is the first study to demonstrate a prognostic potential for 5hmC in PC. Our findings highlight the importance of ERG stratification in PC biomarker studies and suggest that epigenetic mechanisms involving 5hmC are important for the development and/or progression of ERG− PC. Electronic supplementary material The online version of this article (doi:10.1186/s13148-015-0146-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Siri H Strand
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Soren Hoyer
- Department of Histopathology, Aarhus University Hospital, Aarhus, Denmark
| | - Anne-Sofie Lynnerup
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark ; Department of Histopathology, Aarhus University Hospital, Aarhus, Denmark ; Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Christa Haldrup
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Tine Maj Storebjerg
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark ; Department of Histopathology, Aarhus University Hospital, Aarhus, Denmark ; Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Michael Borre
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Torben F Orntoft
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Karina D Sorensen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
24
|
Keil KP, Vezina CM. DNA methylation as a dynamic regulator of development and disease processes: spotlight on the prostate. Epigenomics 2015; 7:413-25. [PMID: 26077429 DOI: 10.2217/epi.15.8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Prostate development, benign hyperplasia and cancer involve androgen and growth factor signaling as well as stromal-epithelial interactions. We review how DNA methylation influences these and related processes in other organ systems such as how proliferation is restricted to specific cell populations during defined temporal windows, how androgens elicit their actions and how cells establish, maintain and remodel DNA methylation in a time and cell specific fashion. We also discuss mechanisms by which hormones and endocrine disrupting chemicals reprogram DNA methylation in the prostate and elsewhere and examine evidence for a reawakening of developmental epigenetic pathways as drivers of prostate cancer and benign prostate hyperplasia.
Collapse
Affiliation(s)
- Kimberly P Keil
- Comparative Biosciences, University of Wisconsin-Madison, 1656 Linden Dr., Madison, WI 53705, USA
| | - Chad M Vezina
- Comparative Biosciences, University of Wisconsin-Madison, 1656 Linden Dr., Madison, WI 53705, USA
| |
Collapse
|
25
|
Struck-Lewicka W, Kordalewska M, Bujak R, Yumba Mpanga A, Markuszewski M, Jacyna J, Matuszewski M, Kaliszan R, Markuszewski MJ. Urine metabolic fingerprinting using LC–MS and GC–MS reveals metabolite changes in prostate cancer: A pilot study. J Pharm Biomed Anal 2015; 111:351-61. [DOI: 10.1016/j.jpba.2014.12.026] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 12/11/2014] [Accepted: 12/13/2014] [Indexed: 01/16/2023]
|
26
|
Chin SP, Marthick JR, West AC, Short AK, Chuckowree J, Polanowski AM, Thomson RJ, Holloway AF, Dickinson JL. Regulation of the ITGA2 gene by epigenetic mechanisms in prostate cancer. Prostate 2015; 75:723-34. [PMID: 25662931 DOI: 10.1002/pros.22954] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 12/01/2014] [Indexed: 11/11/2022]
Abstract
BACKGROUND Integrin alpha2 beta1 (α2 β1 ) plays an integral role in tumour cell invasion, metastasis and angiogenesis, and altered expression of the receptor has been linked to tumour prognosis in several solid tumours. However, the relationship is complex, with both increased and decreased expression associated with different stages of tumour metastases in several tumour types. The ITGA2 gene, which codes for the α2 subunit, was examined to investigate whether a large CpG island associated with its promoter region is involved in the differential expression of ITGA2 observed in prostate cancer. METHODS Bisulphite sequencing of the ITGA2 promoter was used to assess methylation in formalin-fixed paraffin-embedded (FFPE) prostate tumour specimens and prostate cancer cell lines, PC3, 22Rv1 and LNCaP. Changes in ITGA2 mRNA expression were measured using quantitative PCR. ITGA2 functionality was interrogated using cell migration scratch assays and siRNA knockdown experiments. RESULTS Bisulphite sequencing revealed strikingly decreased methylation at key CpG sites within the promoter of tumour samples, when compared with normal prostate tissue. Altered methylation of this CpG island is also associated with differences in expression in the non-invasive LNCaP, and the highly metastatic PC3 and 22Rv1 prostate cancer cell lines. Further bisulphite sequencing confirmed that selected CpGs were highly methylated in LNCaP cells, whilst only low levels of methylation were observed in PC3 and 22Rv1 cells, correlating with ITGA2 transcript levels. Examination of the increased expression of ITGA2 was shown to influence migratory potential via scratch assay in PC3, 22Rv1 and LNCaP cells, and was confirmed by siRNA knockdown experiments. CONCLUSIONS Taken together, our data supports the assertion that epigenetic modification of the ITGA2 promoter is a mechanism by which ITGA2 expression is regulated.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Cell Line, Tumor
- Cell Movement/genetics
- DNA Methylation
- Epigenesis, Genetic
- Gene Expression Regulation, Neoplastic
- Humans
- Integrin alpha5beta1/biosynthesis
- Integrin alpha5beta1/genetics
- Male
- Middle Aged
- Promoter Regions, Genetic
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- RNA, Messenger/chemistry
- RNA, Messenger/genetics
- RNA, Small Interfering/administration & dosage
- RNA, Small Interfering/genetics
- Real-Time Polymerase Chain Reaction
- Sequence Analysis, DNA
Collapse
Affiliation(s)
- Suyin Paulynn Chin
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, 7000, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Bechis SK, Otsetov AG, Ge R, Wang Z, Vangel MG, Wu CL, Tabatabaei S, Olumi AF. Age and Obesity Promote Methylation and Suppression of 5α-Reductase 2: Implications for Personalized Therapy of Benign Prostatic Hyperplasia. J Urol 2015; 194:1031-7. [PMID: 25916673 DOI: 10.1016/j.juro.2015.04.079] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2015] [Indexed: 01/24/2023]
Abstract
PURPOSE In men with symptomatic benign prostatic hyperplasia 5α-reductase inhibitors are a main modality of treatment. More than 30% of men do not respond to the therapeutic effects of 5α-reductase inhibitors. We have found that a third of adult prostate samples do not express 5α-reductase type 2 secondary to epigenetic modifications. We evaluated whether 5α-reductase type 2 expression in benign prostatic hyperplasia specimens from symptomatic men was linked to methylation of the 5α-reductase type 2 gene promoter. We also identified associations with age, obesity, cardiac risk factors and prostate specific antigen. MATERIALS AND METHODS Prostate samples from men undergoing transurethral prostate resection were used. We determined 5α-reductase type 2 protein expression and gene promoter methylation status by common assays. Clinical variables included age, body mass index, hypertension, hyperlipidemia, diabetes, prostate specific antigen and prostate volume. Univariate and multivariate statistical analyses were performed followed by stepwise logistic regression modeling. RESULTS Body mass index and age significantly correlated with methylation of the 5α-reductase type 2 gene promoter (p <0.05) whereas prostate volume, prostate specific antigen or benign prostatic hyperplasia medication did not correlate. Methylation highly correlated with 5α-reductase protein expression (p <0.0001). In a predictive model increasing age and body mass index significantly predicted methylation status and protein expression (p <0.01). CONCLUSIONS Increasing age and body mass index correlate with increased 5α-reductase type 2 gene promoter methylation and decreased protein expression in men with symptomatic benign prostatic hyperplasia. These results highlight the interplay among age, obesity and gene regulation. Our findings suggest an individualized epigenetic signature for symptomatic benign prostatic hyperplasia, which may be important to choose appropriate personalized treatment options.
Collapse
Affiliation(s)
- Seth K Bechis
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alexander G Otsetov
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rongbin Ge
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Zongwei Wang
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mark G Vangel
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Department of Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Chin-Lee Wu
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shahin Tabatabaei
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Aria F Olumi
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
28
|
Abstract
Most recent investigations into cancer etiology have identified a key role played by epigenetics. Specifically, aberrant DNA and histone modifications which silence tumor suppressor genes or promote oncogenes have been demonstrated in multiple cancer models. While the role of epigenetics in several solid tumor cancers such as colorectal cancer are well established, there is emerging evidence that epigenetics also plays a critical role in breast and prostate cancer. In breast cancer, DNA methylation profiles have been linked to hormone receptor status and tumor progression. Similarly in prostate cancer, epigenetic patterns have been associated with androgen receptor status and response to therapy. The regulation of key receptor pathways and activities which affect clinical therapy treatment options by epigenetics renders this field high priority for elucidating mechanisms and potential targets. A new set of methylation arrays are now available to screen epigenetic changes and provide the cutting-edge tools needed to perform such investigations. The role of nutritional interventions affecting epigenetic changes particularly holds promise. Ultimately, determining the causes and outcomes from epigenetic changes will inform translational applications for utilization as biomarkers for risk and prognosis as well as candidates for therapy.
Collapse
Affiliation(s)
- Yanyuan Wu
- Division of Cancer Research and Training, Center to Eliminate Cancer Health Disparities, Department of Internal Medicine, Charles Drew University of Medicine and Science, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA, USA
| | - Marianna Sarkissyan
- Division of Cancer Research and Training, Center to Eliminate Cancer Health Disparities, Department of Internal Medicine, Charles Drew University of Medicine and Science, Los Angeles, CA, USA
| | - Jaydutt V. Vadgama
- Division of Cancer Research and Training, Center to Eliminate Cancer Health Disparities, Department of Internal Medicine, Charles Drew University of Medicine and Science, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA, USA
- Corresponding Author Contact Information: Division of Cancer Research and Training, Center to Eliminate Cancer Health Disparities, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 East 120th Street, Los Angeles, CA 90059, USA. Tele: 323-563-4853. Fax: 323-563-4859 ;
| |
Collapse
|
29
|
Crujeiras AB, Casanueva FF. Obesity and the reproductive system disorders: epigenetics as a potential bridge. Hum Reprod Update 2014; 21:249-61. [DOI: 10.1093/humupd/dmu060] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
30
|
Global DNA hypomethylation in prostate cancer development and progression: a systematic review. Prostate Cancer Prostatic Dis 2014; 18:1-12. [PMID: 25384337 DOI: 10.1038/pcan.2014.45] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 09/10/2014] [Accepted: 09/16/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND The role of global DNA methylation in prostate cancer (PCa) remains largely unknown. Our aim was to summarize evidence on the role of global DNA hypomethylation in PCa development and progression. METHODS We searched PubMed through December 2013 for all studies containing information on global methylation levels in PCa tissue and at least one non-tumor comparison tissue and/or studies reporting association between global methylation levels in PCa tissue and survival, disease recurrence or at least one clinicopathological prognostic factor. We summarized results using non-parametric comparisons and P-value summary methods. RESULTS We included 15 studies in the review: 6 studies with both diagnostic and prognostic information, 5 studies with only diagnostic information and 4 studies with only prognostic information. Quantitative meta-analysis was not possible because of the large heterogeneity in molecular techniques, types of tissues analyzed, aims and study designs. Summary statistical tests showed association of DNA hypomethylation with PCa diagnosis (P<0.006) and prognosis (P<0.001). Restriction to studies assessing 5-methylcytosine or long interspersed nucleotide element-1 revealed results in the same direction. Analyses restricted to specific clinicopathological features showed association with the presence of metastasis and tumor stage in all tests with P<0.03, and no association with Gleason score (all tests P>0.1 except for the weighted Z-test, P=0.05). CONCLUSION DNA hypomethylation was associated with PCa development and progression. However, due to the heterogeneity and small sample sizes of the included studies, along with the possibility of publication bias, this association requires additional assessment.
Collapse
|
31
|
Prognostic DNA methylation markers for prostate cancer. Int J Mol Sci 2014; 15:16544-76. [PMID: 25238417 PMCID: PMC4200823 DOI: 10.3390/ijms150916544] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 09/05/2014] [Accepted: 09/11/2014] [Indexed: 12/14/2022] Open
Abstract
Prostate cancer (PC) is the most commonly diagnosed neoplasm and the third most common cause of cancer-related death amongst men in the Western world. PC is a clinically highly heterogeneous disease, and distinction between aggressive and indolent disease is a major challenge for the management of PC. Currently, no biomarkers or prognostic tools are able to accurately predict tumor progression at the time of diagnosis. Thus, improved biomarkers for PC prognosis are urgently needed. This review focuses on the prognostic potential of DNA methylation biomarkers for PC. Epigenetic changes are hallmarks of PC and associated with malignant initiation as well as tumor progression. Moreover, DNA methylation is the most frequently studied epigenetic alteration in PC, and the prognostic potential of DNA methylation markers for PC has been demonstrated in multiple studies. The most promising methylation marker candidates identified so far include PITX2, C1orf114 (CCDC181) and the GABRE~miR-452~miR-224 locus, in addition to the three-gene signature AOX1/C1orf114/HAPLN3. Several other biomarker candidates have also been investigated, but with less stringent clinical validation and/or conflicting evidence regarding their possible prognostic value available at this time. Here, we review the current evidence for the prognostic potential of DNA methylation markers in PC.
Collapse
|
32
|
Yang SL, Tang KQ, Bai WK, Zhao YW, Shen E, Tao JJ, Hu B. Combined low-frequency ultrasound and microbubble contrast agent for the treatment of benign prostatic hyperplasia. J Endourol 2014; 27:1020-6. [PMID: 23607285 DOI: 10.1089/end.2012.0637] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND AND PURPOSE Benign prostatic hyperplasia is one of the most common conditions in middle-aged and elderly men. The aim of the study was to investigate the treatment effects of low-frequency ultrasound combined with a microbubbles agent on benign prostatic hyperplasia. METHODS Eighteen 7-year-old male beagles with prostatic hyperplasia were randomly divided into six groups (n=3): Control group, 21 kHz ultrasound group, 21 kHz ultrasound and microbubble contrast agent group, 1 MHz ultrasound group, 1 MHz ultrasound and microbubble contrast agent group, and microbubble contrast agent group. The histopathologic damage to prostate cells was assessed via transmission electron microscopy and optical microscopy. The protein expressions of prostate-specific antigen (PSA), inducible nitric oxide synthase (iNOS), and super oxidase dimutase (SOD) were detected by enzyme-linked immunosorbent assay. Levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), blood urea nitrogen (BUN), and creatinine (Cr) were detected by the Biochemistry Automatic Analyzer. RESULTS Significant tissue injury, mitochondria injury, and cell apoptosis were observed in 21 kHz ultrasound and the microbubble contrast agent group. Compared with the control and microbubbles contrast agent groups, the decrease in levels of PSA or increase in levels of iNOs and SOD in the other four groups were statistically significant (P<0.05). The lowest level of PSA and the highest levels of iNOs and SOD were observed in the 21 kHz ultrasound and microbubbles contrast agent group. No significant changes in levels of AST, ALT, BUN, and Cr were observed between the six groups. CONCLUSIONS Our results suggest that lower frequency ultrasound may have a better effect on benign prostatic hyperplasia, and microbubble contrast agent application further strengthens this biological effect.
Collapse
Affiliation(s)
- Shao-ling Yang
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated 6th People's Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
33
|
Novel tools for prostate cancer prognosis, diagnosis, and follow-up. BIOMED RESEARCH INTERNATIONAL 2014; 2014:890697. [PMID: 24877145 PMCID: PMC4024423 DOI: 10.1155/2014/890697] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 04/09/2014] [Indexed: 12/18/2022]
Abstract
Prostate-specific antigen (PSA) is the main diagnostic tool when it comes to prostate cancer but it possesses serious limitations. Therefore, there is an urgent need for more sensitive and specific biomarkers for prostate cancer prognosis and patient follow-up. Recent advances led to the discovery of many novel diagnostic/prognostic techniques and provided us with many worthwhile candidates. This paper briefly reviews the most promising biomarkers with respect to their implementation in screening, early detection, diagnostic confirmation, prognosis, and prediction of therapeutic response or monitoring disease and recurrence; and their use as possible therapeutic targets. This review also examines the possible future directions in the field of prostate cancer marker research.
Collapse
|
34
|
Zhang X, Zheng Z, Yingji S, Kim H, Jin R, Renshu L, Lee DY, Roh MR, Yang S. Downregulation of glutathione peroxidase 3 is associated with lymph node metastasis and prognosis in cervical cancer. Oncol Rep 2014; 31:2587-92. [PMID: 24788695 DOI: 10.3892/or.2014.3152] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 03/21/2014] [Indexed: 11/06/2022] Open
Abstract
Glutathione peroxidase 3 (GPX3) is a member of the glutathione peroxidase family of selenoproteins and is one of the key defensive enzymes against oxidative damages to host cells. Downregulation of GPX3 due to its promoter hypermethylation has been documented in several different types of cancer, indicating that GPX3 functions as a possible tumor suppressor. In the present study, we showed that GPX3 is also significantly downregulated in cervical cancer tissues compared to normal cervical tissues by qRT-PCR analyses and immunohistostainings. GPX3 expression was significantly related to lymph node metastasis and prognosis in cervical cancer patients. Treatment of cervical cancer cells with 5-aza-2'-deoxycytidine restored the expression of GPX3 and methylation-specific PCR (MSP) confirmed the CpG methylation of the GPX3 gene. Our results indicate that promoter methylation is one of the major causes of GPX3 downregulation in cervical cancer and GPX3 could serve as a predictive biomarker for lymph node metastasis and prognosis of cervical cancer.
Collapse
Affiliation(s)
- Xianglan Zhang
- Department of Pathology, Yanbian University Hospital, Yanji, Jilin, P.R. China
| | - Zhenlong Zheng
- Department of Dermatology, Yanbian University Hospital, Yanji, Jilin, P.R. China
| | - Shen Yingji
- Department of Gynecology, Maternal and Child Health Hospital, Dalian, Liaoning, P.R. China
| | - Hyeyeon Kim
- Department of Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Renshun Jin
- Department of Pathology, Yanbian University Hospital, Yanji, Jilin, P.R. China
| | - Li Renshu
- Department of Anesthesia and Pain, Yanbian University Hospital, Yanji, Jilin, P.R. China
| | - Doo Young Lee
- Oral Cancer Research Institute, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| | - Mi Ryung Roh
- Department of Dermatology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sanghwa Yang
- National Creative Research Initiatives Center for Inflammatory Response Modulation, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
35
|
Henrique R, Oliveira AI, Costa VL, Baptista T, Martins AT, Morais A, Oliveira J, Jerónimo C. Epigenetic regulation of MDR1 gene through post-translational histone modifications in prostate cancer. BMC Genomics 2013; 14:898. [PMID: 24344919 PMCID: PMC3878566 DOI: 10.1186/1471-2164-14-898] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 12/11/2013] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Multidrug resistance 1 (MDR1) gene encodes for an ATP binding cassette transporter--P-glycoprotein (P-gp)-- involved in chemoresistance to taxanes. MDR1 promoter methylation is frequent in prostate carcinoma (PCa), suggesting an epigenetic regulation but no functional correlation has been established. We aimed to elucidate the epigenetic mechanisms involved in MDR1 deregulation in PCa. RESULTS MDR1 promoter methylation and P-gp expression were assessed in 121 PCa, 39 high-grade prostatic intraepithelial neoplasia (HGPIN), 28 benign prostatic hyperplasia (BPH) and 10 morphologically normal prostate tissue (NPT) samples, using quantitative methylation specific PCR and immunohistochemistry, respectively. PCa cell lines were exposed to a DNA methyltransferases inhibitor 5-aza-2'deoxycytidine (DAC) and histone deacetylases inhibitor trichostatin A (TSA). Methylation and histone posttranscriptional modifications status were characterized and correlated with mRNA and protein expression. MDR1 promoter methylation levels and frequency significantly increased from NPTs, to HGPIN and to PCa. Conversely, decreased or absent P-gp immunoexpression was observed in HGPIN and PCa, inversely correlating with methylation levels. Exposure to DAC alone did not alter significantly methylation levels, although increased expression was apparent. However, P-gp mRNA and protein re-expression were higher in cell lines exposed to TSA alone or combined with DAC. Accordingly, histone active marks H3Ac, H3K4me2, H3K4me3, H3K9Ac, and H4Ac were increased at the MDR1 promoter after exposure to TSA alone or combined with DAC. CONCLUSION Our data suggests that, in prostate carcinogenesis, MDR1 downregulation is mainly due to histone post-translational modifications. This occurs concomitantly with aberrant promoter methylation, substantiating the association with P-gp decreased expression.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Adult
- Aged
- Aged, 80 and over
- Azacitidine/pharmacology
- Cell Line, Tumor
- CpG Islands
- DNA Methylation
- Epigenesis, Genetic/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Histones/metabolism
- Humans
- Male
- Middle Aged
- Neoplasm Grading
- Neoplasm Staging
- Promoter Regions, Genetic
- Prostate/metabolism
- Prostatic Hyperplasia/genetics
- Prostatic Hyperplasia/metabolism
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Protein Processing, Post-Translational
Collapse
Affiliation(s)
- Rui Henrique
- Cancer Epigenetics Group, Research Center of the Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto, Largo Prof. Abel Salazar 2, 4099-003 Porto, Portugal
| | - Ana Isabel Oliveira
- Cancer Epigenetics Group, Research Center of the Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Vera L Costa
- Cancer Epigenetics Group, Research Center of the Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Tiago Baptista
- Cancer Epigenetics Group, Research Center of the Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Ana Teresa Martins
- Cancer Epigenetics Group, Research Center of the Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - António Morais
- Department of Urology, Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Jorge Oliveira
- Department of Urology, Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Carmen Jerónimo
- Cancer Epigenetics Group, Research Center of the Portuguese Oncology Institute, Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto, Largo Prof. Abel Salazar 2, 4099-003 Porto, Portugal
| |
Collapse
|
36
|
Abstract
Mechanisms postulated to link folate and B12 metabolism with cancer, including genome-wide hypomethylation, gene-specific promoter hypermethylation, and DNA uracil misincorporation, have been observed in prostate tumor cells. However, epidemiological studies of prostate cancer risk, based on dietary intakes and blood levels of folate and vitamin B12 and on folate-pathway gene variants, have generated contradictory findings. In a meta-analysis, circulating concentrations of B12 (seven studies, OR = 1.10; 95% CI 1.01, 1.19; P = 0.002) and (in cohort studies) folate (five studies, OR = 1.18; 95% CI 1.00, 1.40; P = 0.02) were positively associated with an increased risk of prostate cancer. Homocysteine was not associated with risk of prostate cancer (four studies, OR = 0.91; 95% CI 0.69, 1.19; P = 0.5). In a meta-analysis of folate-pathway polymorphisms, MTR 2756A > G (eight studies, OR = 1.06; 95% CI 1.00, 1.12; P = 0.06) and SHMT1 1420C > T (two studies, OR = 1.11; 95% CI 1.00, 1.22; P = 0.05) were positively associated with prostate cancer risk. There were no effects due to any other polymorphisms, including MTHFR 677C > T (12 studies, OR = 1.04; 95% CI 0.97, 1.12; P = 0.3). The positive association of circulating B12 with an increased risk of prostate cancer could be explained by reverse causality. However, given current controversies over mandatory B12 fortification, further research to eliminate a causal role of B12 in prostate cancer initiation and/or progression is required. Meta-analysis does not entirely rule out a positive association of circulating folate with increased prostate cancer risk. As with B12, even a weak positive association would be a significant public health issue, given the high prevalence of prostate cancer and concerns about the potential harms versus benefits of mandatory folic acid fortification.
Collapse
|
37
|
Atala A. Re: EZH2 Oncogenic Activity in Castration-Resistant Prostate Cancer Cells is Polycomb-Independent. J Urol 2013. [DOI: 10.1016/j.juro.2013.03.100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
38
|
Bisphenol A affects androgen receptor function via multiple mechanisms. Chem Biol Interact 2013; 203:556-64. [PMID: 23562765 DOI: 10.1016/j.cbi.2013.03.013] [Citation(s) in RCA: 141] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 03/08/2013] [Accepted: 03/19/2013] [Indexed: 12/21/2022]
Abstract
Bisphenol A (BPA), is a well-known endocrine disruptor compound (EDC) that affects the normal development and function of the female and male reproductive system, however the mechanisms of action remain unclear. To investigate the molecular mechanisms of how BPA may affect ten different nuclear receptors, stable cell lines containing individual nuclear receptor ligand binding domain (LBD)-linked to the β-Gal reporter were examined by a quantitative high throughput screening (qHTS) format in the Tox21 Screening Program of the NIH. The results showed that two receptors, estrogen receptor alpha (ERα) and androgen receptor (AR), are affected by BPA in opposite direction. To confirm the observed effects of BPA on ERα and AR, we performed transient transfection experiments with full-length receptors and their corresponding response elements linked to luciferase reporters. We also included in this study two BPA analogs, bisphenol AF (BPAF) and bisphenol S (BPS). As seen in African green monkey kidney CV1 cells, the present study confirmed that BPA and BPAF act as ERα agonists (half maximal effective concentration EC50 of 10-100 nM) and as AR antagonists (half maximal inhibitory concentration IC50 of 1-2 μM). Both BPA and BPAF antagonized AR function via competitive inhibition of the action of synthetic androgen R1881. BPS with lower estrogenic activity (EC50 of 2.2 μM), did not compete with R1881 for AR binding, when tested at 30 μM. Finally, the effects of BPA were also evaluated in a nuclear translocation assays using EGPF-tagged receptors. Similar to 17β-estradiol (E2) which was used as control, BPA was able to enhance ERα nuclear foci formation but at a 100-fold higher concentration. Although BPA was able to bind AR, the nuclear translocation was reduced. Furthermore, BPA was unable to induce functional foci in the nuclei and is consistent with the transient transfection study that BPA is unable to activate AR.
Collapse
|
39
|
Munfus-McCray D, Cui M, Zhang Z, Gabrielson E, Askin F, Li QK. Comparison of EGFR and KRAS mutations in primary and unpaired metastatic lung adenocarcinoma with potential chemotherapy effect. Hum Pathol 2013; 44:1286-92. [PMID: 23337026 DOI: 10.1016/j.humpath.2012.10.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 10/23/2012] [Accepted: 10/24/2012] [Indexed: 01/03/2023]
Abstract
Several recent studies have suggested that EGFR and KRAS mutations may be different in primary and metastatic tumors. It is also not well studied whether or not conventional chemotherapy has any effect on EGFR or KRAS mutations. In this study, we compared EGFR and KRAS mutations in primary and unrelated metastatic lung adenocarcinomas from retrospectively collected clinical cases. We also examined the potential effect of chemotherapy on EGFR and KRAS mutations in these 2 groups based on available clinical information. Using Johns Hopkins Hospital archives, 379 lung adenocarcinomas with EGFR and KRAS mutational analyses were included. Mutational status was determined by sequencing exons 18 to 21 of EGFR and codons 12 and 13 of KRAS. Clinical information was correlated. The overall mutational rates in primary and metastatic tumors were comparable. In 213 primary tumors, there was no significant difference of EGFR and KRAS mutational rates in the prechemotherapy and postchemotherapy groups (P > .05), whereas in 166 metastatic tumors, EGFR and KRAS mutations were 12.8% and 36.1% in the prechemotherapy group and 27.3% and 18.2% in the postchemotherapy group (P < .05). Although our study is an unpaired study, it suggests that mutational status in metastatic tumors may need to be tested, especially if the patient had chemotherapy before the test. Additional studies are needed to further investigate the mechanism and clinical significance of the findings.
Collapse
Affiliation(s)
- Delicia Munfus-McCray
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA
| | | | | | | | | | | |
Collapse
|
40
|
Treas JN, Tyagi T, Singh KP. Effects of chronic exposure to arsenic and estrogen on epigenetic regulatory genes expression and epigenetic code in human prostate epithelial cells. PLoS One 2012; 7:e43880. [PMID: 22952798 PMCID: PMC3428278 DOI: 10.1371/journal.pone.0043880] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 07/26/2012] [Indexed: 02/05/2023] Open
Abstract
Chronic exposures to arsenic and estrogen are known risk factors for prostate cancer. Though the evidence suggests that exposure to arsenic or estrogens can disrupt normal DNA methylation patterns and histone modifications, the mechanisms by which these chemicals induce epigenetic changes are not fully understood. Moreover, the epigenetic effects of co-exposure to these two chemicals are not known. Therefore, the objective of this study was to evaluate the effects of chronic exposure to arsenic and estrogen, both alone and in combination, on the expression of epigenetic regulatory genes, their consequences on DNA methylation, and histone modifications. Human prostate epithelial cells, RWPE-1, chronically exposed to arsenic and estrogen alone and in combination were used for analysis of epigenetic regulatory genes expression, global DNA methylation changes, and histone modifications at protein level. The result of this study revealed that exposure to arsenic, estrogen, and their combination alters the expression of epigenetic regulatory genes and changes global DNA methylation and histone modification patterns in RWPE-1 cells. These changes were significantly greater in arsenic and estrogen combination treated group than individually treated group. The findings of this study will help explain the epigenetic mechanism of arsenic- and/or estrogen-induced prostate carcinogenesis.
Collapse
Affiliation(s)
- Justin N. Treas
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH), Texas Tech University, Lubbock, Texas, United States of America
| | - Tulika Tyagi
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH), Texas Tech University, Lubbock, Texas, United States of America
| | - Kamaleshwar P. Singh
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH), Texas Tech University, Lubbock, Texas, United States of America
| |
Collapse
|
41
|
Zhao K, Wang W, Yu X, Zhou X, Shi Q, Hu Y. Triptolide-Induced Apoptosis of Androgen-Independent Prostatic Cancer (AIPC) via Modulation of Histone Methylation. Drug Dev Res 2012. [DOI: 10.1002/ddr.21011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Kailiang Zhao
- Department of General Surgery; Renmin Hospital of Wuhan University; Wuhan; China
| | - Weixing Wang
- Department of General Surgery; Renmin Hospital of Wuhan University; Wuhan; China
| | - Xiaohui Yu
- Department of Urology; Renmin Hospital of Wuhan University; Wuhan; China
| | - Xing Zhou
- Department of General Surgery; Renmin Hospital of Wuhan University; Wuhan; China
| | - Qiao Shi
- Department of General Surgery; Renmin Hospital of Wuhan University; Wuhan; China
| | - Yunfei Hu
- Department of Urology; Renmin Hospital of Wuhan University; Wuhan; China
| |
Collapse
|
42
|
Lee C, Zhang Q, Zi X, Dash A, Soares MB, Rahmatpanah F, Jia Z, McClelland M, Mercola D. TGF-β mediated DNA methylation in prostate cancer. Transl Androl Urol 2012; 1:78-88. [PMID: 25133096 PMCID: PMC4131550 DOI: 10.3978/j.issn.2223-4683.2012.05.06] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 05/04/2012] [Indexed: 12/15/2022] Open
Abstract
Almost all tumors harbor a defective negative feedback loop of signaling by transforming growth factor-β (TGF-β). Epigenetic mechanisms of gene regulation, including DNA methylation, are fundamental to normal cellular function and also play a major role in carcinogenesis. Recent evidence demonstrated that TGF-β signaling mediates cancer development and progression. Many key events in TGF-β signaling in cancer included auto-induction of TGF-β1 and increased expression of DNA methyltransferases (DNMTs), suggesting that DNA methylation plays a significant role in cancer development and progression. In this review, we performed an extensive survey of the literature linking TGF-β signaling to DNA methylation in prostate cancer. It appeared that almost all DNA methylated genes detected in prostate cancer are directly or indirectly related to TGF-β signaling. This knowledge has provided a basis for our future directions of prostate cancer research and strategies for prevention and therapy for prostate cancer.
Collapse
|
43
|
Positive and negative regulation of prostate stem cell antigen expression by Yin Yang 1 in prostate epithelial cell lines. PLoS One 2012; 7:e35570. [PMID: 22536409 PMCID: PMC3334921 DOI: 10.1371/journal.pone.0035570] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Accepted: 03/20/2012] [Indexed: 01/12/2023] Open
Abstract
Prostate cancer is influenced by epigenetic modification of genes involved in cancer development and progression. Increased expression of Prostate Stem Cell Antigen (PSCA) is correlated with development of malignant human prostate cancer, while studies in mouse models suggest that decreased PSCA levels promote prostate cancer metastasis. These studies suggest that PSCA has context-dependent functions, and could be differentially regulated during tumor progression. In the present study, we identified the multi-functional transcription factor Yin Yang 1 (YY1) as a modulator of PSCA expression in prostate epithelial cell lines. Increased YY1 levels are observed in prostatic intraepithelial neoplasia (PIN) and advanced disease. We show that androgen-mediated up-regulation of PSCA in prostate epithelial cell lines is dependent on YY1. We identified two direct YY1 binding sites within the PSCA promoter, and showed that the upstream site inhibited, while the downstream site, proximal to the androgen-responsive element, stimulated PSCA promoter activity. Thus, changes in PSCA expression levels in prostate cancer may at least partly be affected by cellular levels of YY1. Our results also suggest multiple roles for YY1 in prostate cancer which may contribute to disease progression by modulation of genes such as PSCA.
Collapse
|
44
|
Chen Z, Wang L, Wang Q, Li W. Histone modifications and chromatin organization in prostate cancer. Epigenomics 2012; 2:551-60. [PMID: 21318127 DOI: 10.2217/epi.10.31] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Epigenetic mechanisms, including histone modifications, nucleosomal remodeling and chromosomal looping, contribute to the onset and progression of prostate cancer. Recent technical advances significantly increase our understanding of the genome-wide epigenetic regulation of gene expression in prostate cancer. Aberrant genomic distribution and global level of histone modifications, nucleosome repositioning at the gene promoter and enhancer regions, as well as androgen receptor-mediated chromosomal looping may lead to the silencing of tumor suppressor genes and the activation of proto-oncogenes. In addition, androgen receptor-induced chromosomal looping facilitates recurrent gene fusion in prostate cancer. Studies in epigenetic regulation have translational implications in the identification of new biomarkers and the development of new therapies in prostate cancer.
Collapse
Affiliation(s)
- Zhong Chen
- Department of Molecular & Cellular Biochemistry & Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | |
Collapse
|
45
|
Epigenetic biomarkers in prostate cancer: Current and future uses. Cancer Lett 2012; 342:248-56. [PMID: 22391123 DOI: 10.1016/j.canlet.2012.02.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 02/10/2012] [Accepted: 02/11/2012] [Indexed: 12/18/2022]
Abstract
Epigenome alterations are characteristic of nearly all human malignancies and include changes in DNA methylation, histone modifications and microRNAs (miRNAs). However, what induces these epigenetic alterations in cancer is largely unknown and their mechanistic role in prostate tumorigenesis is just beginning to be evaluated. Identification of the epigenetic modifications involved in the development and progression of prostate cancer will not only identify novel therapeutic targets but also prognostic and diagnostic markers. This review will focus on the use of epigenetic modifications as biomarkers for prostate cancer.
Collapse
|
46
|
Role of Polymorphic Variants of MTR Gene A2756G and SHMT1 Gene C1420T in the Development of Prostatic Cancer in Residents of the Western Siberian Region of Russia. Bull Exp Biol Med 2012; 152:466-9. [DOI: 10.1007/s10517-012-1554-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
47
|
MTHFR polymorphism and the risk of prostate cancer: a meta-analysis of case–control studies. Prostate Cancer Prostatic Dis 2012; 15:244-9. [DOI: 10.1038/pcan.2012.5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
48
|
Li D, Tian T, Guo C, Ren J, Yan L, Liu H, Xu Z. No association of the MTHFR gene A1298C polymorphism with the risk of prostate cancer: A meta-analysis. Exp Ther Med 2012; 3:493-498. [PMID: 22969917 DOI: 10.3892/etm.2012.445] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 11/24/2011] [Indexed: 11/05/2022] Open
Abstract
Various studies have demonstrated that the 5,10-methylenetetrahydrofolate reductase (MTHFR) gene polymorphism contributes to the risk of prostate cancer, while other studies have provided conflicting findings. In the present study, we carried out a comprehensive meta-analysis with the aim of determining whether there is a significant association of the MTHFR gene A1298C polymorphism with the susceptibility of prostate cancer. Studies on the MTHFR gene A1298C polymorphism and prostate cancer were retrieved using the electronic PubMed database without any restriction on language through Aug 21, 2011. Data were abstracted by a standardized protocol. Crude odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to estimate the strength of association. The analyses were conducted with Review Manager software version 4.2. Nine case-control studies were identified, including 2,723 prostate cancer patients and 3,442 controls. Overall, no significant associations were found between the MTHFR gene A1298C polymorphism and prostate cancer (codominant models: CC vs. AA, OR=1.03, 95% CI 0.79-1.34, P=0.84; AC vs. AA, OR=1.04, 95% CI 0.93-1.16, P=0.46; dominant model: AC + CC vs. AA, OR=1.04, 95% CI 0.94-1.15, P=0.48; recessive model: CC vs. AC + AA, OR=1.02, 95% CI 0.76-1.35, P=0.91; allele model: C vs. A, OR=1.04, 95% CI 0.90-1.19, P=0.61). Similarly, in the subgroup analyses by DNA source, ethnicity, control source, pathological stage and Hardy-Weinberg equilibrium, no significant associations were observed. Our meta-analysis suggests that the MTHFR gene A1298C polymorphism is not associated with the risk of prostate cancer.
Collapse
Affiliation(s)
- Dawei Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong 250012
| | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
The androgen receptor (AR) is a key transcriptional regulator and therapeutic target in prostate cancer. During androgen deprivation therapy to treat metastatic prostate cancer, surviving cells acquire increased AR signaling through a variety of mechanisms, one of which is enhanced interactions with AR coactivators. One recently identified AR-specific coregulator expressed only in human and nonhuman primates is the melanoma antigen gene protein-A11 (MAGE-11). MAGE-11 increases AR transcriptional activity through direct interactions with AR and other coactivators, and its levels increase during prostate cancer progression to castration-recurrent growth. The MAGE-11 gene is located at Xq28 on the human X chromosome as part of an X-linked MAGE gene family of cancer-testis antigens. MAGE-11 stabilizes AR when androgen levels are low, and functions in a transcriptional hub to promote AR-mediated gene activation. The evolutionary development and organization of the MAGE-11 gene within the cancer-testis antigen family suggests that MAGE-11 provides a gain-of-function to AR among primates in both normal physiology and cancer, and may serve as a therapeutic target in the treatment of advanced prostate cancer.
Collapse
Affiliation(s)
- Elizabeth M Wilson
- Laboratories for Reproductive Biology, Lineberger Comprehensive Cancer Center, and the Departments of Pediatrics, and Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
50
|
Ho E, Beaver LM, Williams DE, Dashwood RH. Dietary factors and epigenetic regulation for prostate cancer prevention. Adv Nutr 2011; 2:497-510. [PMID: 22332092 PMCID: PMC3226387 DOI: 10.3945/an.111.001032] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The role of epigenetic alterations in various human chronic diseases has gained increasing attention and has resulted in a paradigm shift in our understanding of disease susceptibility. In the field of cancer research, e.g., genetic abnormalities/mutations historically were viewed as primary underlying causes; however, epigenetic mechanisms that alter gene expression without affecting DNA sequence are now recognized as being of equal or greater importance for oncogenesis. Methylation of DNA, modification of histones, and interfering microRNA (miRNA) collectively represent a cadre of epigenetic elements dysregulated in cancer. Targeting the epigenome with compounds that modulate DNA methylation, histone marks, and miRNA profiles represents an evolving strategy for cancer chemoprevention, and these approaches are starting to show promise in human clinical trials. Essential micronutrients such as folate, vitamin B-12, selenium, and zinc as well as the dietary phytochemicals sulforaphane, tea polyphenols, curcumin, and allyl sulfur compounds are among a growing list of agents that affect epigenetic events as novel mechanisms of chemoprevention. To illustrate these concepts, the current review highlights the interactions among nutrients, epigenetics, and prostate cancer susceptibility. In particular, we focus on epigenetic dysregulation and the impact of specific nutrients and food components on DNA methylation and histone modifications that can alter gene expression and influence prostate cancer progression.
Collapse
Affiliation(s)
- Emily Ho
- Linus Pauling Institute, Department of Nutrition and Exercise Sciences, Oregon State University, Corvallis, OR, USA.
| | - Laura M. Beaver
- Linus Pauling Institute,Department of Nutrition and Exercise Sciences, and
| | - David E. Williams
- Linus Pauling Institute,Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR
| | - Roderick H. Dashwood
- Linus Pauling Institute,Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR
| |
Collapse
|