1
|
Constâncio V, Lobo J, Sequeira JP, Henrique R, Jerónimo C. Prostate cancer epigenetics - from pathophysiology to clinical application. Nat Rev Urol 2025:10.1038/s41585-024-00991-8. [PMID: 39820138 DOI: 10.1038/s41585-024-00991-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 01/19/2025]
Abstract
Prostate cancer is a multifactorial disease influenced by various molecular features. Over the past decades, epigenetics, which is the study of changes in gene expression without altering the DNA sequence, has been recognized as a major driver of this disease. In the past 50 years, advancements in technological tools to characterize the epigenome have highlighted crucial roles of epigenetic mechanisms throughout the entire spectrum of prostate cancer, from initiation to progression, including localized disease, metastatic dissemination, castration resistance and neuroendocrine transdifferentiation. Substantial advances in the understanding of epigenetic mechanisms in the pathophysiology of prostate cancer have been carried out, but translating preclinical achievements into clinical practice remains challenging. Ongoing research and biomarker-oriented clinical trials are expected to increase the likelihood of successfully integrating epigenetics into prostate cancer clinical management.
Collapse
Affiliation(s)
- Vera Constâncio
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, Portugal
- Doctoral Program in Biomedical Sciences, ICBAS - School of Medicine & Biomedical Sciences, University of Porto (ICBAS-UP), Porto, Portugal
| | - João Lobo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre Raquel Seruca (Porto.CCC), Porto, Portugal
- Department of Pathology and Molecular Immunology, ICBAS - School of Medicine & Biomedical Sciences, University of Porto, Porto, Portugal
| | - José Pedro Sequeira
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, Portugal
- Doctoral Program in Biomedical Sciences, ICBAS - School of Medicine & Biomedical Sciences, University of Porto (ICBAS-UP), Porto, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre Raquel Seruca (Porto.CCC), Porto, Portugal
- Department of Pathology and Molecular Immunology, ICBAS - School of Medicine & Biomedical Sciences, University of Porto, Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, Portugal.
- Department of Pathology and Molecular Immunology, ICBAS - School of Medicine & Biomedical Sciences, University of Porto, Porto, Portugal.
| |
Collapse
|
2
|
Cacciatore A, Shinde D, Musumeci C, Sandrini G, Guarrera L, Albino D, Civenni G, Storelli E, Mosole S, Federici E, Fusina A, Iozzo M, Rinaldi A, Pecoraro M, Geiger R, Bolis M, Catapano CV, Carbone GM. Epigenome-wide impact of MAT2A sustains the androgen-indifferent state and confers synthetic vulnerability in ERG fusion-positive prostate cancer. Nat Commun 2024; 15:6672. [PMID: 39107274 PMCID: PMC11303763 DOI: 10.1038/s41467-024-50908-7] [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: 04/11/2023] [Accepted: 07/25/2024] [Indexed: 08/09/2024] Open
Abstract
Castration-resistant prostate cancer (CRPC) is a frequently occurring disease with adverse clinical outcomes and limited therapeutic options. Here, we identify methionine adenosyltransferase 2a (MAT2A) as a critical driver of the androgen-indifferent state in ERG fusion-positive CRPC. MAT2A is upregulated in CRPC and cooperates with ERG in promoting cell plasticity, stemness and tumorigenesis. RNA, ATAC and ChIP-sequencing coupled with histone post-translational modification analysis by mass spectrometry show that MAT2A broadly impacts the transcriptional and epigenetic landscape. MAT2A enhances H3K4me2 at multiple genomic sites, promoting the expression of pro-tumorigenic non-canonical AR target genes. Genetic and pharmacological inhibition of MAT2A reverses the transcriptional and epigenetic remodeling in CRPC models and improves the response to AR and EZH2 inhibitors. These data reveal a role of MAT2A in epigenetic reprogramming and provide a proof of concept for testing MAT2A inhibitors in CRPC patients to improve clinical responses and prevent treatment resistance.
Collapse
MESH Headings
- Male
- Humans
- Transcriptional Regulator ERG/genetics
- Transcriptional Regulator ERG/metabolism
- Methionine Adenosyltransferase/genetics
- Methionine Adenosyltransferase/metabolism
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/pathology
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic/drug effects
- Epigenesis, Genetic/drug effects
- Animals
- Androgens/metabolism
- Epigenome
- Mice
- Histones/metabolism
- Receptors, Androgen/metabolism
- Receptors, Androgen/genetics
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Enhancer of Zeste Homolog 2 Protein/metabolism
- Enhancer of Zeste Homolog 2 Protein/genetics
- Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors
Collapse
Affiliation(s)
- Alessia Cacciatore
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Dheeraj Shinde
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Carola Musumeci
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Giada Sandrini
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
- Swiss Institute of Bioinformatics, Bioinformatics Core Unit, 6500, Bellinzona, Switzerland
| | - Luca Guarrera
- Istituto di Ricerche Farmacologiche "Mario Negri" IRCCS, 20156, Milano, Italy
| | - Domenico Albino
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Gianluca Civenni
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Elisa Storelli
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Simone Mosole
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Elisa Federici
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Alessio Fusina
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Marta Iozzo
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Andrea Rinaldi
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Matteo Pecoraro
- Institute for Research in Biomedicine (IRB), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Roger Geiger
- Institute for Research in Biomedicine (IRB), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Marco Bolis
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
- Istituto di Ricerche Farmacologiche "Mario Negri" IRCCS, 20156, Milano, Italy
| | - Carlo V Catapano
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland
| | - Giuseppina M Carbone
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), 6500, Bellinzona, Switzerland.
| |
Collapse
|
3
|
Oladipo EK, Olufemi SE, Adediran DA, Adejumo IO, Jimah EM, Oloke JK, Udekwu CC, Ogunwobi OO. Epigenetic modifications in solid tumor metastasis in people of African ancestry. Front Oncol 2024; 14:1325614. [PMID: 38450190 PMCID: PMC10915648 DOI: 10.3389/fonc.2024.1325614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 01/18/2024] [Indexed: 03/08/2024] Open
Abstract
This review focuses on the critical role of epigenetic modifications in solid tumor metastasis, particularly in people of African ancestry. Epigenetic alterations, such as DNA methylation, histone modifications, alterations in non-coding RNAs, and mRNA methylation, significantly influence gene expression, contributing to cancer development and progression. Despite the primary focus on populations of European, American, and Asian descent in most cancer research, this work emphasizes the importance of studying the unique genetic and epigenetic landscapes of African populations for a more inclusive approach in understanding and treating cancer. Insights from this review have the potential to pave the way for the development of effective, tailored treatments, and provide a richer resource for understanding cancer progression and metastasis. Specific focus was placed on the role of DNA methylation, histone modifications, non-coding RNAs, and mRNA methylation in solid tumor metastasis, including how these modifications contribute to the regulation of tumor suppressor genes and oncogenes, influence cellular pathways and signaling, and interact with the immune system. Moreover, this review elaborates on the development of epigenetic-targeted therapeutic strategies and the current advances in this field, highlighting the promising applications of these therapies in improving outcomes for African ancestry populations disproportionately affected by certain types of cancer. Nevertheless, this work acknowledges the challenges that lie ahead, particularly the under-representation of African populations in cancer genomic and epigenomic studies and the technical complications associated with detecting subtle epigenetic modifications. Emphasis is placed on the necessity for more inclusive research practices, the development of more robust and sensitive methods for detecting and interpreting epigenetic changes, and the understanding of the interplay between genetic and epigenetic variations. The review concludes with an optimistic outlook on the future of epigenetic research in People of African ancestry, urging the concerted efforts of researchers, clinicians, funding agencies, and policymakers to extend the benefits of this research to all populations.
Collapse
Affiliation(s)
- Elijah Kolawole Oladipo
- Genomics Unit, Helix Biogen Institute, Ogbomoso, Oyo, Nigeria
- Laboratory of Molecular Biology, Immunology and Bioinformatics, Adeleke University, Ede, Osun State, Nigeria
| | - Seun Elijah Olufemi
- Genomics Unit, Helix Biogen Institute, Ogbomoso, Oyo, Nigeria
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria
| | - Daniel Adewole Adediran
- Genomics Unit, Helix Biogen Institute, Ogbomoso, Oyo, Nigeria
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria
| | | | | | - Julius Kola Oloke
- Genomics Unit, Helix Biogen Institute, Ogbomoso, Oyo, Nigeria
- Department of Natural Sciences, Precious Cornerstone University, Ibadan, Nigeria
| | - Chinedum C. Udekwu
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States
| | - Olorunseun O. Ogunwobi
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
4
|
Zaalberg A, Pottendorfer E, Zwart W, Bergman AM. It Takes Two to Tango: The Interplay between Prostate Cancer and Its Microenvironment from an Epigenetic Perspective. Cancers (Basel) 2024; 16:294. [PMID: 38254784 PMCID: PMC10813511 DOI: 10.3390/cancers16020294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/28/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
Prostate cancer is the second most common cancer in men worldwide and is associated with high morbidity and mortality. Consequently, there is an urgent unmet need for novel treatment avenues. In addition to somatic genetic alterations, deviations in the epigenetic landscape of cancer cells and their tumor microenvironment (TME) are critical drivers of prostate cancer initiation and progression. Unlike genomic mutations, epigenetic modifications are potentially reversible. Therefore, the inhibition of aberrant epigenetic modifications represents an attractive and exciting novel treatment strategy for castration-resistant prostate cancer patients. Moreover, drugs targeting the epigenome also exhibit synergistic interactions with conventional therapeutics by directly enhancing their anti-tumorigenic properties by "priming" the tumor and tumor microenvironment to increase drug sensitivity. This review summarizes the major epigenetic alterations in prostate cancer and its TME, and their involvement in prostate tumorigenesis, and discusses the impact of epigenome-targeted therapies.
Collapse
Affiliation(s)
- Anniek Zaalberg
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; (A.Z.); (E.P.)
| | - Elisabeth Pottendorfer
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; (A.Z.); (E.P.)
| | - Wilbert Zwart
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; (A.Z.); (E.P.)
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Oncode Institute
| | - Andries M. Bergman
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; (A.Z.); (E.P.)
- Division of Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
5
|
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
|
6
|
Johnson RP, Ratnacaram CK, Kumar L, Jose J. Combinatorial approaches of nanotherapeutics for inflammatory pathway targeted therapy of prostate cancer. Drug Resist Updat 2022; 64:100865. [PMID: 36099796 DOI: 10.1016/j.drup.2022.100865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 12/24/2022]
Abstract
Prostate cancer (PC) is the most prevalent male urogenital cancer worldwide. PC patients presenting an advanced or metastatic cancer succumb to the disease, even after therapeutic interventions including radiotherapy, surgery, androgen deprivation therapy (ADT), and chemotherapy. One of the hallmarks of PC is evading immune surveillance and chronic inflammation, which is a major challenge towards designing effective therapeutic formulations against PC. Chronic inflammation in PC is often characterized by tumor microenvironment alterations, epithelial-mesenchymal transition and extracellular matrix modifications. The inflammatory events are modulated by reactive nitrogen and oxygen species, inflammatory cytokines and chemokines. Major signaling pathways in PC includes androgen receptor, PI3K and NF-κB pathways and targeting these inter-linked pathways poses a major therapeutic challenge. Notably, many conventional treatments are clinically unsuccessful, due to lack of targetability and poor bioavailability of the therapeutics, untoward toxicity and multidrug resistance. The past decade witnessed an advancement of nanotechnology as an excellent therapeutic paradigm for PC therapy. Modern nanovectorization strategies such as stimuli-responsive and active PC targeting carriers offer controlled release patterns and superior anti-cancer effects. The current review initially describes the classification, inflammatory triggers and major inflammatory pathways of PC, various PC treatment strategies and their limitations. Subsequently, recent advancement in combinatorial nanotherapeutic approaches, which target PC inflammatory pathways, and the mechanism of action are discussed. Besides, the current clinical status and prospects of PC homing nanovectorization, and major challenges to be addressed towards the advancement PC therapy are also addressed.
Collapse
Affiliation(s)
- Renjith P Johnson
- Polymer Nanobiomaterial Research Laboratory, Nanoscience and Microfluidics Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Chandrahas Koumar Ratnacaram
- Cell Signaling and Cancer Biology Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Lalit Kumar
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka 576 104, India
| | - Jobin Jose
- NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Mangalore 575018, India.
| |
Collapse
|
7
|
Treas J, Roy P, Singh KP. Chronic coexposure to arsenic and estrogen potentiates genotoxic estrogen metabolic pathway and hypermethylation of DNA glycosylase MBD4 in human prostate epithelial cells. Prostate 2022; 82:1273-1283. [PMID: 35747940 DOI: 10.1002/pros.24401] [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: 12/24/2021] [Revised: 05/14/2022] [Accepted: 06/01/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Previously we reported that arsenic and estrogen cause synergistic effects in the neoplastic transformation of human prostate epithelial cells. In addition to receptor-mediated pathways, DNA-reactive estrogen metabolites have also been shown to play a critical role in mutagenicity and carcinogenicity. Both estrogen and arsenic are known prostate carcinogens, however, the effect of coexposure to these two chemicals on genes involved in estrogen metabolism is not known. Therefore, the objective of this study was to evaluate the role of arsenic and estrogen coexposure on the expression of estrogen receptors and estrogen metabolism-associated genes. Earlier, we also reported the synergistic effect of arsenic and estrogen on decreased expression of MBD4 genes that play an important role in DNA repair through its DNA glycosylase activity. To further understand the mechanism, the promoter methylation of this gene was also analyzed. METHODS Total RNA and protein were isolated from RWPE-1 human prostate epithelial cells that were coexposed to arsenic and estrogen for a chronic duration (6 months). The expression of estrogen receptors, estrogen metabolism associated phase I genes (CYP 1A1, 1A2, 3A4, and 1B1) and phase II gene catechol-O-methyltransferase (COMT), as well as antioxidant MnSOD, were analyzed either at the RNA level by quantitative reverse transcriptase-polymerase chain reaction or at the protein level by western blot. Promoter methylation of MBD4 was analyzed by pyrosequencing. RESULTS Expression of MnSOD and phase I genes that convert E2 into genotoxic metabolites 2-OH-E2 and 4-OH-E2 were significantly increased, whereas the expression of phase II gene COMT that detoxifies estrogen metabolites was significantly decreased in arsenic and estrogen coexposed cells. MBD4 promoter was hypermethylated in arsenic and estrogen coexposed cells. Coexposure to arsenic and estrogen has synergistic effects on the expression of these genes as well as in MBD4 promoter hypermethylation. CONCLUSIONS These novel findings suggest that coexposure to arsenic and estrogen acts synergistically in the activation of not only the estrogen receptors but also the genes associated with genotoxic estrogen metabolism and epigenetic inactivation of DNA glycosylase MBD4. Together, these genetic and epigenetic aberrations provide the molecular basis for the potentiation of carcinogenicity of arsenic and estrogen coexposure in prostate epithelial cells.
Collapse
Affiliation(s)
- Justin Treas
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Priti Roy
- Department of Internal Medicine, University of California San Francisco, San Francisco, California, USA
| | - Kamaleshwar P Singh
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH), Texas Tech University, Lubbock, Texas, USA
| |
Collapse
|
8
|
Dufresne S, Guéritat J, Wong CP, Isanejad A, Ho E, Serna E, Gomez-Cabrera MC, Rebillard A. Exercise training as a modulator of epigenetic events in prostate tumors. Prostate Cancer Prostatic Dis 2022; 25:119-122. [PMID: 34007020 DOI: 10.1038/s41391-021-00380-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/18/2021] [Accepted: 04/28/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND Exercise is increasingly recognized as an effective strategy to improve cancer prevention and prognosis. Several biological mechanisms mediating these benefits have been proposed, but the role of epigenetics remains largely unknown. Since epigenetics is highly susceptible to lifestyle factors, we hypothesized that exercise could affect the epigenome landscape in cancer tissues. METHODS Rats implanted with AT1 prostate tumors were randomized to either control or exercise training. microRNA expression, DNA methylation and histone acetylation were analyzed in the tumor tissue. RESULTS MiR-27a-5p appeared to be differently expressed between sedentary and trained rats. Furthermore, exercise increased global DNA methylation and decreased DNA methyltransferases mRNA expression in the tumor tissue. Histone acetylation however remained unaltered. CONCLUSION Overall, exercise might reverse some of the cancer-related epigenetic alterations in the prostate tumor tissue.
Collapse
Affiliation(s)
| | | | - Carmen P Wong
- School of Biological & Population Health Sciences, College of Public Health & Human Sciences, 211 Milam Hall, Oregon State University, Corvallis, OR, USA
- Linus Pauling Institute, Oregon State University, 307 Linus Pauling Science Center, Corvallis, OR, USA
| | | | - Emily Ho
- School of Biological & Population Health Sciences, College of Public Health & Human Sciences, 211 Milam Hall, Oregon State University, Corvallis, OR, USA
- Linus Pauling Institute, Oregon State University, 307 Linus Pauling Science Center, Corvallis, OR, USA
- Moore Family Center for Whole Grain Foods, Nutrition & Preventive Health, Oregon State University, Corvallis, OR, USA
| | - Eva Serna
- Freshage Research Group, Department of Physiology, University of Valencia, CIBERFES, INCLIVA, Valencia, Spain
| | - Marie-Carmen Gomez-Cabrera
- Freshage Research Group, Department of Physiology, University of Valencia, CIBERFES, INCLIVA, Valencia, Spain
| | | |
Collapse
|
9
|
Pardo JC, Ruiz de Porras V, Gil J, Font A, Puig-Domingo M, Jordà M. Lipid Metabolism and Epigenetics Crosstalk in Prostate Cancer. Nutrients 2022; 14:851. [PMID: 35215499 PMCID: PMC8874497 DOI: 10.3390/nu14040851] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/27/2022] [Accepted: 02/14/2022] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (PCa) is the most commonly diagnosed malignant neoplasm in men in the Western world. Localized low-risk PCa has an excellent prognosis thanks to effective local treatments; however, despite the incorporation of new therapeutic strategies, metastatic PCa remains incurable mainly due to disease heterogeneity and the development of resistance to therapy. The mechanisms underlying PCa progression and therapy resistance are multiple and include metabolic reprogramming, especially in relation to lipid metabolism, as well as epigenetic remodelling, both of which enable cancer cells to adapt to dynamic changes in the tumour. Interestingly, metabolism and epigenetics are interconnected. Metabolism can regulate epigenetics through the direct influence of metabolites on epigenetic processes, while epigenetics can control metabolism by directly or indirectly regulating the expression of metabolic genes. Moreover, epidemiological studies suggest an association between a high-fat diet, which can alter the availability of metabolites, and PCa progression. Here, we review the alterations of lipid metabolism and epigenetics in PCa, before focusing on the mechanisms that connect them. We also discuss the influence of diet in this scenario. This information may help to identify prognostic and predictive biomarkers as well as targetable vulnerabilities.
Collapse
Affiliation(s)
- Juan C. Pardo
- Department of Medical Oncology, Catalan Institute of Oncology, University Hospital Germans Trias i Pujol, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain; (J.C.P.); (A.F.)
- Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (B·ARGO), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain;
| | - Vicenç Ruiz de Porras
- Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (B·ARGO), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain;
- Germans Trias i Pujol Research Institute (IGTP), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain; (J.G.); (M.P.-D.)
| | - Joan Gil
- Germans Trias i Pujol Research Institute (IGTP), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain; (J.G.); (M.P.-D.)
- Department of Endocrinology and Medicine, CIBERER U747, ISCIII, Research Center for Pituitary Diseases, Hospital Sant Pau, IIB-SPau, Universitat Autònoma de Barcelona, 08041 Barcelona, Spain
| | - Albert Font
- Department of Medical Oncology, Catalan Institute of Oncology, University Hospital Germans Trias i Pujol, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain; (J.C.P.); (A.F.)
- Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (B·ARGO), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain;
| | - Manel Puig-Domingo
- Germans Trias i Pujol Research Institute (IGTP), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain; (J.G.); (M.P.-D.)
- Department of Endocrinology and Nutrition, University Germans Trias i Pujol Hospital, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain
- Department of Medicine, Autonomous University of Barcelona (UAB), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain
| | - Mireia Jordà
- Germans Trias i Pujol Research Institute (IGTP), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain; (J.G.); (M.P.-D.)
| |
Collapse
|
10
|
Inflammation, Fibrosis and Cancer: Mechanisms, Therapeutic Options and Challenges. Cancers (Basel) 2022; 14:cancers14030552. [PMID: 35158821 PMCID: PMC8833582 DOI: 10.3390/cancers14030552] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/15/2022] [Accepted: 01/17/2022] [Indexed: 01/09/2023] Open
Abstract
Uncontrolled inflammation is a salient factor in multiple chronic inflammatory diseases and cancers. In this review, we provided an in-depth analysis of the relationships and distinctions between uncontrolled inflammation, fibrosis and cancers, while emphasizing the challenges and opportunities of developing novel therapies for the treatment and/or management of these diseases. We described how drug delivery systems, combination therapy and the integration of tissue-targeted and/or pathways selective strategies could overcome the challenges of current agents for managing and/or treating chronic inflammatory diseases and cancers. We also recognized the value of the re-evaluation of the disease-specific roles of multiple pathways implicated in the pathophysiology of chronic inflammatory diseases and cancers-as well as the application of data from single-cell RNA sequencing in the success of future drug discovery endeavors.
Collapse
|
11
|
Roggero CM, Esser V, Duan L, Rice AM, Ma S, Raj GV, Rosen MK, Liu ZP, Rizo J. Poly-glutamine-dependent self-association as a potential mechanism for regulation of androgen receptor activity. PLoS One 2022; 17:e0258876. [PMID: 34986150 PMCID: PMC8730435 DOI: 10.1371/journal.pone.0258876] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/13/2021] [Indexed: 01/01/2023] Open
Abstract
The androgen receptor (AR) plays a central role in prostate cancer. Development of castration resistant prostate cancer (CRPC) requires androgen-independent activation of AR, which involves its large N-terminal domain (NTD) and entails extensive epigenetic changes depending in part on histone lysine demethylases (KDMs) that interact with AR. The AR-NTD is rich in low-complexity sequences, including a polyQ repeat. Longer polyQ sequences were reported to decrease transcriptional activity and to protect against prostate cancer, although they can lead to muscular atrophy. However, the molecular mechanisms underlying these observations are unclear. Using NMR spectroscopy, here we identify weak interactions between the AR-NTD and the KDM4A catalytic domain, and between the AR ligand-binding domain and a central KDM4A region that also contains low-complexity sequences. We also show that the AR-NTD can undergo liquid-liquid phase separation in vitro, with longer polyQ sequences phase separating more readily. Moreover, longer polyQ sequences hinder nuclear localization in the absence of hormone and increase the propensity for formation of AR-containing puncta in the nucleus of cells treated with dihydrotestosterone. These results lead us to hypothesize that polyQ-dependent liquid-liquid phase separation may provide a mechanism to decrease the transcriptional activity of AR, potentially opening new opportunities to design effective therapies against CRPC and muscular atrophy.
Collapse
Affiliation(s)
- Carlos M. Roggero
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Victoria Esser
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Lingling Duan
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Allyson M. Rice
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Shihong Ma
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Ganesh V. Raj
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Michael K. Rosen
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Zhi-Ping Liu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Josep Rizo
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| |
Collapse
|
12
|
Li C, Wang W, Xie SS, Ma WX, Fan QW, Chen Y, He Y, Wang JN, Yang Q, Li HD, Jin J, Liu MM, Meng XM, Wen JG. The Programmed Cell Death of Macrophages, Endothelial Cells, and Tubular Epithelial Cells in Sepsis-AKI. Front Med (Lausanne) 2021; 8:796724. [PMID: 34926535 PMCID: PMC8674574 DOI: 10.3389/fmed.2021.796724] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Sepsis is a systemic inflammatory response syndrome caused by infection, following with acute injury to multiple organs. Sepsis-induced acute kidney injury (AKI) is currently recognized as one of the most severe complications related to sepsis. The pathophysiology of sepsis-AKI involves multiple cell types, including macrophages, vascular endothelial cells (ECs) and renal tubular epithelial cells (TECs), etc. More significantly, programmed cell death including apoptosis, necroptosis and pyroptosis could be triggered by sepsis in these types of cells, which enhances AKI progress. Moreover, the cross-talk and connections between these cells and cell death are critical for better understanding the pathophysiological basis of sepsis-AKI. Mitochondria dysfunction and oxidative stress are traditionally considered as the leading triggers of programmed cell death. Recent findings also highlight that autophagy, mitochondria quality control and epigenetic modification, which interact with programmed cell death, participate in the damage process in sepsis-AKI. The insightful understanding of the programmed cell death in sepsis-AKI could facilitate the development of effective treatment, as well as preventive methods.
Collapse
Affiliation(s)
- Chao Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Wei Wang
- Anhui Province Key Laboratory of Genitourinary Diseases, Department of Urology and Institute of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Shuai-Shuai Xie
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Wen-Xian Ma
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Qian-Wen Fan
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Ying Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Yuan He
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jia-Nan Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Qin Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Hai-di Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Juan Jin
- Key Laboratory of Anti-inflammatory and Immunopharmacology (Ministry of Education), Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Ming-Ming Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jia-Gen Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-Inflammatory of Immune Medicines (Ministry of Education), Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| |
Collapse
|
13
|
Ruiz de la Cruz M, de la Cruz Montoya AH, Rojas Jiménez EA, Martínez Gregorio H, Díaz Velásquez CE, Paredes de la Vega J, de la Cruz Hernández-Hernández F, Vaca Paniagua F. Cis-Acting Factors Causing Secondary Epimutations: Impact on the Risk for Cancer and Other Diseases. Cancers (Basel) 2021; 13:cancers13194807. [PMID: 34638292 PMCID: PMC8508567 DOI: 10.3390/cancers13194807] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/09/2021] [Accepted: 08/15/2021] [Indexed: 12/25/2022] Open
Abstract
Epigenetics affects gene expression and contributes to disease development by alterations known as epimutations. Hypermethylation that results in transcriptional silencing of tumor suppressor genes has been described in patients with hereditary cancers and without pathogenic variants in the coding region of cancer susceptibility genes. Although somatic promoter hypermethylation of these genes can occur in later stages of the carcinogenic process, constitutional methylation can be a crucial event during the first steps of tumorigenesis, accelerating tumor development. Primary epimutations originate independently of changes in the DNA sequence, while secondary epimutations are a consequence of a mutation in a cis or trans-acting factor. Secondary epimutations have a genetic basis in cis of the promoter regions of genes involved in familial cancers. This highlights epimutations as a novel carcinogenic mechanism whose contribution to human diseases is underestimated by the scarcity of the variants described. In this review, we provide an overview of secondary epimutations and present evidence of their impact on cancer. We propose the necessity for genetic screening of loci associated with secondary epimutations in familial cancer as part of prevention programs to improve molecular diagnosis, secondary prevention, and reduce the mortality of these diseases.
Collapse
Affiliation(s)
- Miguel Ruiz de la Cruz
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla 54090, Mexico; (M.R.d.l.C.); (E.A.R.J.); (H.M.G.); (C.E.D.V.); (J.P.d.l.V.)
- Avenida Instituto Politécnico Nacional # 2508, Colonia San Pedro Zacatenco, Delegación Gustavo A. Madero, C.P. Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico;
| | | | - Ernesto Arturo Rojas Jiménez
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla 54090, Mexico; (M.R.d.l.C.); (E.A.R.J.); (H.M.G.); (C.E.D.V.); (J.P.d.l.V.)
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla 54090, Mexico;
| | - Héctor Martínez Gregorio
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla 54090, Mexico; (M.R.d.l.C.); (E.A.R.J.); (H.M.G.); (C.E.D.V.); (J.P.d.l.V.)
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla 54090, Mexico;
| | - Clara Estela Díaz Velásquez
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla 54090, Mexico; (M.R.d.l.C.); (E.A.R.J.); (H.M.G.); (C.E.D.V.); (J.P.d.l.V.)
| | - Jimena Paredes de la Vega
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla 54090, Mexico; (M.R.d.l.C.); (E.A.R.J.); (H.M.G.); (C.E.D.V.); (J.P.d.l.V.)
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla 54090, Mexico;
| | - Fidel de la Cruz Hernández-Hernández
- Avenida Instituto Politécnico Nacional # 2508, Colonia San Pedro Zacatenco, Delegación Gustavo A. Madero, C.P. Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico;
| | - Felipe Vaca Paniagua
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla 54090, Mexico; (M.R.d.l.C.); (E.A.R.J.); (H.M.G.); (C.E.D.V.); (J.P.d.l.V.)
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla 54090, Mexico;
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México 14080, Mexico
- Correspondence: ; Tel.: +52-55-5623-1333 (ext. 39788)
| |
Collapse
|
14
|
Conteduca V, Hess J, Yamada Y, Ku SY, Beltran H. Epigenetics in prostate cancer: clinical implications. Transl Androl Urol 2021; 10:3104-3116. [PMID: 34430414 PMCID: PMC8350251 DOI: 10.21037/tau-20-1339] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/21/2021] [Indexed: 12/18/2022] Open
Abstract
Epigenetic alterations, including changes in DNA methylation, histone modifications and nucleosome remodeling, result in abnormal gene expression patterns that contribute to prostate tumor initiation and continue to evolve during the course of disease progression. Epigenetic modifications are responsible for silencing tumor-suppressor genes, activating oncogenic drivers, and driving therapy resistance and thus have emerged as promising targets for antineoplastic therapy in prostate cancer. In this review, we discuss the role of epigenetics in prostate cancer with a particular emphasis on clinical implications. We review how epigenetic regulators crosstalk with critical biological pathways, including androgen receptor signaling, and how these interactions dynamically control prostate cancer transcriptional profiles. Because of their potentially reversible nature, restoration of a "normal" epigenome could provide a basis for innovative therapeutic strategies in prostate cancer. We highlight how particular epigenetic alterations are emerging as potential diagnostic and prognostic biomarkers and/or targets for the treatment of advanced prostate cancer.
Collapse
Affiliation(s)
- Vincenza Conteduca
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori "Dino Amadori" (IRST) IRCCS, Meldola, Italy
| | - Judy Hess
- Weill Cornell Medicine, New York, NY, USA
| | - Yasutaka Yamada
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Sheng-Yu Ku
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Himisha Beltran
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Wang K, Li Y, Qiang T, Chen J, Wang X. Role of epigenetic regulation in myocardial ischemia/reperfusion injury. Pharmacol Res 2021; 170:105743. [PMID: 34182132 DOI: 10.1016/j.phrs.2021.105743] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/09/2021] [Accepted: 06/23/2021] [Indexed: 12/30/2022]
Abstract
Nowadays acute myocardial infarction (AMI) is a serious cardiovascular disease threatening the human life and health worldwide. The most effective treatment is to quickly restore coronary blood flow through revascularization. However, timely revascularization may lead to reperfusion injury, thereby reducing the clinical benefits of revascularization. At present, no effective treatment is available for myocardial ischemia/reperfusion injury. Emerging evidence indicates that epigenetic regulation is closely related to the pathogenesis of myocardial ischemia/reperfusion injury, indicating that epigenetics may serve as a novel therapeutic target to ameliorate or prevent ischemia/reperfusion injury. This review aimed to briefly summarize the role of histone modification, DNA methylation, noncoding RNAs, and N6-methyladenosine (m6A) methylation in myocardial ischemia/reperfusion injury, with a view to providing new methods and ideas for the research and treatment of myocardial ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Keyan Wang
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China,; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai 201203, China
| | - Yiping Li
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China,; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai 201203, China
| | - Tingting Qiang
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China,; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai 201203, China
| | - Jie Chen
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China,; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai 201203, China
| | - Xiaolong Wang
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China,; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai 201203, China.
| |
Collapse
|
16
|
Liu J, Liu J, Duan S, Liu L, Zhang G, Peng X. Reprogrammed Epigenetic Landscape-Prophesied Functions of Bioactive Polysaccharides in Alleviating Diseases: A Pilot Study of DNA Methylome Remodeling in Astragalus Polysaccharide (APS)-Improved Osteoporosis in a Rat Model. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:15449-15459. [PMID: 33320666 DOI: 10.1021/acs.jafc.0c06483] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
DNA methylation is an epigenetic event that plays critical roles in the pathogenesis, progression, and treatment of human diseases. In this study, we investigated the epigenetic mechanisms for Astragalus polysaccharide (APS)-improved osteoporosis in a rat model. The results showed that APS significantly changed the DNA methylome in colonic epithelia with great efficiency. Gene set enrichment analysis (GSEA) based on differentially methylated sites (DMSs) revealed that APS caused promoter DNA methylation changes of genes associated with calcium homeostasis, osteoclast/osteoblast balance, Wnt signaling, and hormone-related processes. Further analysis showed high consistency of APS-induced gene methylomic changes in colonic epithelia and its effects on diabetes, virus infection, and wound healing, which had been reported already. Moreover, we suggested new functions and the involved mechanisms of APS in heart disease, neurological disorder, reproductive problem, and olfactory dysfunction. In this study, we offered epigenetic mechanisms for APS-improved osteoporosis. More importantly, we proposed and proved a reliable method to explore the beneficial effects of bioactive polysaccharides by studying DNA methylation changes at nonfocal sites. We firmly believed the promising prospects of this method for its great efficiency, rapidness, and economy in exploring possible beneficial or therapeutic effects of functional macromolecules with one single experiment.
Collapse
Affiliation(s)
- Junsheng Liu
- Department of Food Science and Engineering, Jinan University, No. 601 West Huangpu Avenue, Guangzhou, Guangdong 510632, P. R. China
| | - Jun Liu
- Department of Food Science and Engineering, Jinan University, No. 601 West Huangpu Avenue, Guangzhou, Guangdong 510632, P. R. China
| | - Shan Duan
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510642, P. R. China
| | - Liu Liu
- Department of Food Science and Engineering, Jinan University, No. 601 West Huangpu Avenue, Guangzhou, Guangdong 510632, P. R. China
| | - Guangwen Zhang
- Department of Food Science and Engineering, Jinan University, No. 601 West Huangpu Avenue, Guangzhou, Guangdong 510632, P. R. China
| | - Xichun Peng
- Department of Food Science and Engineering, Jinan University, No. 601 West Huangpu Avenue, Guangzhou, Guangdong 510632, P. R. China
| |
Collapse
|
17
|
Burke B, Eden C, Perez C, Belshoff A, Hart S, Plaza-Rojas L, Delos Reyes M, Prajapati K, Voelkel-Johnson C, Henry E, Gupta G, Guevara-Patiño J. Inhibition of Histone Deacetylase (HDAC) Enhances Checkpoint Blockade Efficacy by Rendering Bladder Cancer Cells Visible for T Cell-Mediated Destruction. Front Oncol 2020; 10:699. [PMID: 32500025 PMCID: PMC7243798 DOI: 10.3389/fonc.2020.00699] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
Inhibitory checkpoint blockade therapy is an immunomodulatory strategy that results in the restoration of T cell functions, and its efficacy depends on the recognition of tumor cells for destruction. Considering the factors at play, one could propose that anti-tumor responses will not occur if tumor cells are immunologically invisible to T cells. In this study, we tested a strategy based on the modulation of cancer cell's immunovisibility through HDAC inhibition. In a model (heterotopic and orthotopic) of mouse urothelial bladder cancer, we demonstrated that the use of intratumoral or intravesical HDACi in combination with systemic anti-PD-1 was effective at inducing curative responses with durable anti-tumor immunity capable of preventing tumor growth at a distal site. Mechanistically, we determined that protective responses were dependent on CD8 cells, but not NK cells. Of significance, in an in vitro human model, we found that fully activated T cells fail at killing bladder cancer cells unless tumor cells were pretreated with HDACi. Complementary to this observation, we found that HDACi cause gene deregulation, that results in the upregulation of genes responsible for mediating immunorecognition, NKG2D ligands and HSP70. Taken together, these data indicate that HDAC inhibition results in the elimination of the tumor cell's “invisibility cloak” that prevents T cells from recognizing and killing them. Finally, as checkpoint blockade therapy moves into the adjuvant setting, its combined use with locally administrated HDACi represents a new approach to be included in our current therapeutic treatment toolbox.
Collapse
Affiliation(s)
- Brianna Burke
- Department of Surgery and Cancer Biology, Loyola University Chicago, Chicago, IL, United States
| | - Catherine Eden
- Department of Urology, Loyola University Medical Center, Maywood, IL, United States
| | - Cynthia Perez
- Department of Surgery and Cancer Biology, Loyola University Chicago, Chicago, IL, United States
| | - Alex Belshoff
- Department of Urology, Loyola University Medical Center, Maywood, IL, United States
| | - Spencer Hart
- Department of Urology, Loyola University Medical Center, Maywood, IL, United States
| | - Lourdes Plaza-Rojas
- Department of Surgery and Cancer Biology, Loyola University Chicago, Chicago, IL, United States
| | - Michael Delos Reyes
- Department of Surgery and Cancer Biology, Loyola University Chicago, Chicago, IL, United States
| | - Kushal Prajapati
- Department of Surgery and Cancer Biology, Loyola University Chicago, Chicago, IL, United States
| | - Christina Voelkel-Johnson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Elizabeth Henry
- Department of Oncology, Loyola University Medical Center, Maywood, IL, United States
| | - Gopal Gupta
- Department of Surgery and Cancer Biology, Loyola University Chicago, Chicago, IL, United States.,Department of Urology, Loyola University Medical Center, Maywood, IL, United States
| | - José Guevara-Patiño
- Department of Surgery and Cancer Biology, Loyola University Chicago, Chicago, IL, United States
| |
Collapse
|
18
|
Richa S, Dey P, Park C, Yang J, Son JY, Park JH, Lee SH, Ahn MY, Kim IS, Moon HR, Kim HS. A New Histone Deacetylase Inhibitor, MHY4381, Induces Apoptosis via Generation of Reactive Oxygen Species in Human Prostate Cancer Cells. Biomol Ther (Seoul) 2020; 28:184-194. [PMID: 31476841 PMCID: PMC7059815 DOI: 10.4062/biomolther.2019.074] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/13/2019] [Accepted: 07/23/2019] [Indexed: 01/14/2023] Open
Abstract
Histone deacetylase (HDAC) inhibitors represent a novel class of anticancer agents, which can be used to inhibit cell proliferation and induce apoptosis in several types of cancer cells. In this study, we investigated the anticancer activity of MHY4381, a newly synthesized HDAC inhibitor, against human prostate cancer cell lines and compared its efficacy with that of suberoylanilide hydroxamic acid (SAHA), a well-known HDAC inhibitor. We assessed cell viability, apoptosis, cell cycle regulation, and other biological effects in the prostate cancer cells. We also evaluated a possible mechanism of MHY4381 on the apoptotic cell death pathway. The IC50 value of MHY4381 was lower in DU145 cells (IC50=0.31 µM) than in LNCaP (IC50=0.85 µM) and PC-3 cells (IC50=5.23 µM). In addition, the IC50 values of MHY4381 measured in this assay were significantly lower than those of SAHA against prostate cancer cell lines. MHY4381 increased the levels of acetylated histones H3 and H4 and reduced the expression of HDAC proteins in the prostate cancer cell lines. MHY4381 increased G2/M phase arrest in DU145 cells, and G1 arrest in LNCaP cells. It also activated reactive oxygen species (ROS) generation, which induced apoptosis in the DU145 and LNCaP cells by increasing the ratio of Bax/Bcl-2 and releasing cytochrome c into the cytoplasm. Our results indicated that MHY4381 preferentially results in antitumor effects in DU145 and LNCaP cells via mitochondria-mediated apoptosis and ROS-facilitated cell death pathway, and therefore can be used as a promising prostate cancer therapeutic.
Collapse
Affiliation(s)
- Sachan Richa
- School of Pharmacy, Sungkyunkwan University, Suwon 16419
| | - Prasanta Dey
- School of Pharmacy, Sungkyunkwan University, Suwon 16419
| | - Chaeun Park
- College of Pharmacy, Pusan National University, Busan 46241
| | - Jungho Yang
- College of Pharmacy, Pusan National University, Busan 46241
| | - Ji Yeon Son
- School of Pharmacy, Sungkyunkwan University, Suwon 16419
| | - Jae Hyeon Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419
| | - Su Hyun Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419
| | - Mee-Young Ahn
- Major in Pharmaceutical Engineering, Division of Bioindustry, College of Medical and Life Sciences, Silla University, Busan 46958,
Republic of Korea
| | - In Su Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419
| | | | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419
| |
Collapse
|
19
|
Patel PG, Wessel T, Kawashima A, Okello JBA, Jamaspishvili T, Guérard KP, Lee L, Lee AYW, How NE, Dion D, Scarlata E, Jackson CL, Boursalie S, Sack T, Dunn R, Moussa M, Mackie/ K, Ellis A, Marra E, Chin J, Siddiqui K, Hetou K, Pickard LA, Arthur-Hayward V, Bauman G, Chevalier S, Brimo F, Boutros PC, Lapointe PhD J, Bartlett JMS, Gooding RJ, Berman DM. A three-gene DNA methylation biomarker accurately classifies early stage prostate cancer. Prostate 2019; 79:1705-1714. [PMID: 31433512 DOI: 10.1002/pros.23895] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/29/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND We identify and validate accurate diagnostic biomarkers for prostate cancer through a systematic evaluation of DNA methylation alterations. MATERIALS AND METHODS We assembled three early prostate cancer cohorts (total patients = 699) from which we collected and processed over 1300 prostatectomy tissue samples for DNA extraction. Using real-time methylation-specific PCR, we measured normalized methylation levels at 15 frequently methylated loci. After partitioning sample sets into independent training and validation cohorts, classifiers were developed using logistic regression, analyzed, and validated. RESULTS In the training dataset, DNA methylation levels at 7 of 15 genomic loci (glutathione S-transferase Pi 1 [GSTP1], CCDC181, hyaluronan, and proteoglycan link protein 3 [HAPLN3], GSTM2, growth arrest-specific 6 [GAS6], RASSF1, and APC) showed large differences between cancer and benign samples. The best binary classifier was the GAS6/GSTP1/HAPLN3 logistic regression model, with an area under these curves of 0.97, which showed a sensitivity of 94%, and a specificity of 93% after external validation. CONCLUSION We created and validated a multigene model for the classification of benign and malignant prostate tissue. With false positive and negative rates below 7%, this three-gene biomarker represents a promising basis for more accurate prostate cancer diagnosis.
Collapse
Affiliation(s)
- Palak G Patel
- Department of Pathology & Molecular Medicine, Queen's University, Kingston, Ontario, Canada
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
| | - Thomas Wessel
- Life Sciences Group, Thermo Fisher Scientific, Waltham, Massachusetts
| | - Atsunari Kawashima
- Department of Pathology & Molecular Medicine, Queen's University, Kingston, Ontario, Canada
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - John B A Okello
- Department of Pathology & Molecular Medicine, Queen's University, Kingston, Ontario, Canada
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
- Cardiac Genome Clinic, Ted Rogers Centre for Heart Research, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Tamara Jamaspishvili
- Department of Pathology & Molecular Medicine, Queen's University, Kingston, Ontario, Canada
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
| | - Karl-Philippe Guérard
- Division of Urology, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Québec, Canada
| | - Laura Lee
- Ontario Institute for Cancer Research (OICR), Toronto, Ontario, Canada
| | - Anna Ying-Wah Lee
- Ontario Institute for Cancer Research (OICR), Toronto, Ontario, Canada
| | - Nathan E How
- Department of Pathology & Molecular Medicine, Queen's University, Kingston, Ontario, Canada
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
| | - Dan Dion
- Ontario Institute for Cancer Research (OICR), Toronto, Ontario, Canada
| | - Eleonora Scarlata
- Division of Urology, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Québec, Canada
| | - Chelsea L Jackson
- Department of Pathology & Molecular Medicine, Queen's University, Kingston, Ontario, Canada
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
| | - Suzanne Boursalie
- Department of Pathology & Molecular Medicine, Queen's University, Kingston, Ontario, Canada
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
| | - Tanya Sack
- Department of Pathology & Molecular Medicine, Queen's University, Kingston, Ontario, Canada
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
| | - Rachel Dunn
- Department of Pathology & Molecular Medicine, Queen's University, Kingston, Ontario, Canada
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
| | - Madeleine Moussa
- Division of Surgical Pathology, Departmant of Pathology and Laboratory Medicine, London Health Sciences Centre, London, Ontario, Canada
| | - Karen Mackie/
- Division of Surgical Pathology, Departmant of Pathology and Laboratory Medicine, London Health Sciences Centre, London, Ontario, Canada
| | - Audrey Ellis
- Division of Surgical Pathology, Departmant of Pathology and Laboratory Medicine, London Health Sciences Centre, London, Ontario, Canada
| | - Elizabeth Marra
- Division of Surgical Pathology, Departmant of Pathology and Laboratory Medicine, London Health Sciences Centre, London, Ontario, Canada
| | - Joseph Chin
- Department of Surgery (Urology), London Health Sciences Centre, London, ON, Canada
| | - Khurram Siddiqui
- Department of Surgery (Urology), London Health Sciences Centre, London, ON, Canada
| | - Khalil Hetou
- Department of Surgery (Urology), London Health Sciences Centre, London, ON, Canada
| | | | | | - Glenn Bauman
- Division of Radiation Oncology, London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
- Department of Physics and Astronomy, University of Western Ontario, London, Ontario, Canada
| | - Simone Chevalier
- Division of Urology, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Québec, Canada
| | - Fadi Brimo
- Department of Pathology, McGill University Health Center and McGill University, Montreal, Québec, Canada
| | - Paul C Boutros
- Ontario Institute for Cancer Research (OICR), Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Departments of Urology and Human Genetics, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | - Jacques Lapointe PhD
- Division of Urology, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Québec, Canada
| | - John M S Bartlett
- Diagnostic Development, Ontario Institute for Cancer Research (OICR), Toronto, Ontario, Canada
| | - Robert J Gooding
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
- Department of Physics, Engineering Physics & Astronomy, Queen's University, Kingston, Ontario, Canada
| | - David M Berman
- Department of Pathology & Molecular Medicine, Queen's University, Kingston, Ontario, Canada
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
20
|
Civenni G, Albino D, Shinde D, Vázquez R, Merulla J, Kokanovic A, Mapelli SN, Carbone GM, Catapano CV. Transcriptional Reprogramming and Novel Therapeutic Approaches for Targeting Prostate Cancer Stem Cells. Front Oncol 2019; 9:385. [PMID: 31143708 PMCID: PMC6521702 DOI: 10.3389/fonc.2019.00385] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/25/2019] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer is the most common malignancy in men and the second cause of cancer-related deaths in western countries. Despite the progress in the treatment of localized prostate cancer, there is still lack of effective therapies for the advanced forms of the disease. Most patients with advanced prostate cancer become resistant to androgen deprivation therapy (ADT), which remains the main therapeutic option in this setting, and progress to lethal metastatic castration-resistant prostate cancer (mCRPC). Current therapies for prostate cancer preferentially target proliferating, partially differentiated, and AR-dependent cancer cells that constitute the bulk of the tumor mass. However, the subpopulation of tumor-initiating or tumor-propagating stem-like cancer cells is virtually resistant to the standard treatments causing tumor relapse at the primary or metastatic sites. Understanding the pathways controlling the establishment, expansion and maintenance of the cancer stem cell (CSC) subpopulation is an important step toward the development of more effective treatment for prostate cancer, which might enable ablation or exhaustion of CSCs and prevent treatment resistance and disease recurrence. In this review, we focus on the impact of transcriptional regulators on phenotypic reprogramming of prostate CSCs and provide examples supporting the possibility of inhibiting maintenance and expansion of the CSC pool in human prostate cancer along with the currently available methodological approaches. Transcription factors are key elements for instructing specific transcriptional programs and inducing CSC-associated phenotypic changes implicated in disease progression and treatment resistance. Recent studies have shown that interfering with these processes causes exhaustion of CSCs with loss of self-renewal and tumorigenic capability in prostate cancer models. Targeting key transcriptional regulators in prostate CSCs is a valid therapeutic strategy waiting to be tested in clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Carlo V. Catapano
- Institute of Oncology (IOR), Università della Svizzera Italiana, Bellinzona, Switzerland
| |
Collapse
|
21
|
Braadland PR, Urbanucci A. Chromatin reprogramming as an adaptation mechanism in advanced prostate cancer. Endocr Relat Cancer 2019; 26:R211-R235. [PMID: 30844748 DOI: 10.1530/erc-18-0579] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 02/15/2019] [Indexed: 12/13/2022]
Abstract
Tumor evolution is based on the ability to constantly mutate and activate different pathways under the selective pressure of targeted therapies. Epigenetic alterations including those of the chromatin structure are associated with tumor initiation, progression and drug resistance. Many cancers, including prostate cancer, present enlarged nuclei, and chromatin appears altered and irregular. These phenotypic changes are likely to result from epigenetic dysregulation. High-throughput sequencing applied to bulk samples and now to single cells has made it possible to study these processes in unprecedented detail. It is therefore timely to review the impact of chromatin relaxation and increased DNA accessibility on prostate cancer growth and drug resistance, and their effects on gene expression. In particular, we focus on the contribution of chromatin-associated proteins such as the bromodomain-containing proteins to chromatin relaxation. We discuss the consequence of this for androgen receptor transcriptional activity and briefly summarize wider gain-of-function effects on other oncogenic transcription factors and implications for more effective prostate cancer treatment.
Collapse
Affiliation(s)
- Peder Rustøen Braadland
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alfonso Urbanucci
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
- Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, Forskningsparken, University of Oslo, Oslo, Norway
| |
Collapse
|
22
|
Allas L, Boumédiene K, Baugé C. Epigenetic dynamic during endochondral ossification and articular cartilage development. Bone 2019; 120:523-532. [PMID: 30296494 DOI: 10.1016/j.bone.2018.10.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/03/2018] [Accepted: 10/04/2018] [Indexed: 12/23/2022]
Abstract
Within the last decade epigenetics has emerged as fundamental regulator of numerous cellular processes, including those orchestrating embryonic and fetal development. As such, epigenetic factors play especially crucial roles in endochondral ossification, the process by which bone tissue is created, as well during articular cartilage formation. In this review, we summarize the recent discoveries that characterize how DNA methylation, histone post-translational modifications and non-coding RNA (e.g., miRNA and lcnRNA) epigenetically regulate endochondral ossification and chondrogenesis.
Collapse
Affiliation(s)
- Lyess Allas
- Normandie Univ, UNICAEN, EA7451 BioConnecT, Caen, France
| | | | | |
Collapse
|
23
|
Tang J, Zhuang S. Histone acetylation and DNA methylation in ischemia/reperfusion injury. Clin Sci (Lond) 2019; 133:597-609. [PMID: 30804072 PMCID: PMC7470454 DOI: 10.1042/cs20180465] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 01/25/2019] [Accepted: 02/11/2019] [Indexed: 12/17/2022]
Abstract
Ischemic/reperfusion (I/R) injury causes a series of serious clinical problems associated with high morbidity and mortality in various disorders, such as acute kidney injury (AKI), myocardial infarction, ischemic stroke, circulatory arrest, and peripheral vascular disease. The pathophysiology and pathogenesis of I/R injury is complex and multifactorial. Recent studies have revealed that epigenetic regulation is critically involved in the pathogenesis of I/R-induced tissue injury. In this review, we will sum up recent advances on the modification, regulation, and implication of histone modifications and DNA methylation in I/R injury-induced organ dysfunction. Understandings of I/R-induced epigenetic alterations and regulations will aid in the development of potential therapeutics.
Collapse
Affiliation(s)
- Jinhua Tang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, U.S.A
| |
Collapse
|
24
|
Rubicz R, Zhao S, Geybels M, Wright JL, Kolb S, Klotzle B, Bibikova M, Troyer D, Lance R, Ostrander EA, Feng Z, Fan JB, Stanford JL. DNA methylation profiles in African American prostate cancer patients in relation to disease progression. Genomics 2019; 111:10-16. [PMID: 26902887 PMCID: PMC4992660 DOI: 10.1016/j.ygeno.2016.02.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Revised: 12/02/2015] [Accepted: 02/18/2016] [Indexed: 12/25/2022]
Abstract
This study examined whether differential DNA methylation is associated with clinical features of more aggressive disease at diagnosis and prostate cancer recurrence in African American men, who are more likely to die from prostate cancer than other populations. Tumor tissues from 76 African Americans diagnosed with prostate cancer who had radical prostatectomy as their primary treatment were profiled for epigenome-wide DNA methylation levels. Long-term follow-up identified 19 patients with prostate cancer recurrence. Twenty-three CpGs were differentially methylated (FDR q≤0.25, mean methylation difference≥0.10) in patients with vs. without recurrence, including CpGs in GCK, CDKL2, PRDM13, and ZFR2. Methylation differences were also observed between men with metastatic-lethal prostate cancer vs. no recurrence (five CpGs), regional vs. local pathological stage (two CpGs), and higher vs. lower tumor aggressiveness (one CpG). These results indicate that differentially methylated CpG sites identified in tumor tissues of African American men may contribute to prostate cancer aggressiveness.
Collapse
Affiliation(s)
- Rohina Rubicz
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, United States.
| | - Shanshan Zhao
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Milan Geybels
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Jonathan L Wright
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, United States; Department of Urology, University of Washington School of Medicine, Seattle, WA, United States
| | - Suzanne Kolb
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | | | | | - Dean Troyer
- Department of Pathology, Eastern Virginia Medical School, Norfolk, VA, United States; Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Raymond Lance
- Department of Urology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Elaine A Ostrander
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, United States
| | - Ziding Feng
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - Janet L Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, United States; Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, United States
| |
Collapse
|
25
|
Smits M, Mehra N, Sedelaar M, Gerritsen W, Schalken JA. Molecular biomarkers to guide precision medicine in localized prostate cancer. Expert Rev Mol Diagn 2018. [PMID: 28635333 DOI: 10.1080/14737159.2017.1345627] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Major advances through tumor profiling technologies, that include next-generation sequencing, epigenetic, proteomic and transcriptomic methods, have been made in primary prostate cancer, providing novel biomarkers that may guide precision medicine in the near future. Areas covered: The authors provided an overview of novel molecular biomarkers in tissue, blood and urine that may be used as clinical tools to assess prognosis, improve selection criteria for active surveillance programs, and detect disease relapse early in localized prostate cancer. Expert commentary: Active surveillance (AS) in localized prostate cancer is an accepted strategy in patients with very low-risk prostate cancer. Many more patients may benefit from watchful waiting, and include patients of higher clinical stage and grade, however selection criteria have to be optimized and early recognition of transformation from localized to lethal disease has to be improved by addition of molecular biomarkers. The role of non-invasive biomarkers is challenging the need for repeat biopsies, commonly performed at 1 and 4 years in men under AS programs.
Collapse
Affiliation(s)
- Minke Smits
- a Department of Urology and Oncology , Radboud Universiteit , Nijmegen , The Netherlands
| | - Niven Mehra
- a Department of Urology and Oncology , Radboud Universiteit , Nijmegen , The Netherlands
| | - Michiel Sedelaar
- a Department of Urology and Oncology , Radboud Universiteit , Nijmegen , The Netherlands
| | - Winald Gerritsen
- a Department of Urology and Oncology , Radboud Universiteit , Nijmegen , The Netherlands
| | - Jack A Schalken
- a Department of Urology and Oncology , Radboud Universiteit , Nijmegen , The Netherlands
| |
Collapse
|
26
|
Urbanucci A, Barfeld SJ, Kytölä V, Itkonen HM, Coleman IM, Vodák D, Sjöblom L, Sheng X, Tolonen T, Minner S, Burdelski C, Kivinummi KK, Kohvakka A, Kregel S, Takhar M, Alshalalfa M, Davicioni E, Erho N, Lloyd P, Karnes RJ, Ross AE, Schaeffer EM, Vander Griend DJ, Knapp S, Corey E, Feng FY, Nelson PS, Saatcioglu F, Knudsen KE, Tammela TLJ, Sauter G, Schlomm T, Nykter M, Visakorpi T, Mills IG. Androgen Receptor Deregulation Drives Bromodomain-Mediated Chromatin Alterations in Prostate Cancer. Cell Rep 2018; 19:2045-2059. [PMID: 28591577 DOI: 10.1016/j.celrep.2017.05.049] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 04/01/2017] [Accepted: 05/12/2017] [Indexed: 12/17/2022] Open
Abstract
Global changes in chromatin accessibility may drive cancer progression by reprogramming transcription factor (TF) binding. In addition, histone acetylation readers such as bromodomain-containing protein 4 (BRD4) have been shown to associate with these TFs and contribute to aggressive cancers including prostate cancer (PC). Here, we show that chromatin accessibility defines castration-resistant prostate cancer (CRPC). We show that the deregulation of androgen receptor (AR) expression is a driver of chromatin relaxation and that AR/androgen-regulated bromodomain-containing proteins (BRDs) mediate this effect. We also report that BRDs are overexpressed in CRPCs and that ATAD2 and BRD2 have prognostic value. Finally, we developed gene stratification signature (BROMO-10) for bromodomain response and PC prognostication, to inform current and future trials with drugs targeting these processes. Our findings provide a compelling rational for combination therapy targeting bromodomains in selected patients in which BRD-mediated TF binding is enhanced or modified as cancer progresses.
Collapse
Affiliation(s)
- Alfonso Urbanucci
- Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, Forskningsparken, University of Oslo, 21 0349 Oslo, Norway; Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway.
| | - Stefan J Barfeld
- Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, Forskningsparken, University of Oslo, 21 0349 Oslo, Norway
| | - Ville Kytölä
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Tampere University of Technology, 33520 Tampere, Finland
| | - Harri M Itkonen
- Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, Forskningsparken, University of Oslo, 21 0349 Oslo, Norway
| | - Ilsa M Coleman
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Daniel Vodák
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0424 Oslo, Norway
| | - Liisa Sjöblom
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Fimlab Laboratories, Tampere University Hospital, 33520 Tampere, Finland
| | - Xia Sheng
- Department of Biosciences, University of Oslo, 0316 Oslo, Norway
| | - Teemu Tolonen
- Department of Pathology, Fimlab Laboratories, Tampere University Hospital, 33520 Tampere, Finland
| | - Sarah Minner
- University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Christoph Burdelski
- General, Visceral and Thoracic Surgery Department and Clinic, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Kati K Kivinummi
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Tampere University of Technology, 33520 Tampere, Finland
| | - Annika Kohvakka
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Fimlab Laboratories, Tampere University Hospital, 33520 Tampere, Finland
| | - Steven Kregel
- Department of Surgery - Section of Urology, University of Chicago, Chicago, IL 60637, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI 48109-0940, USA
| | - Mandeep Takhar
- Research and Development, GenomeDx Biosciences, Vancouver, BC V6B 1B8, Canada
| | - Mohammed Alshalalfa
- Research and Development, GenomeDx Biosciences, Vancouver, BC V6B 1B8, Canada
| | - Elai Davicioni
- Research and Development, GenomeDx Biosciences, Vancouver, BC V6B 1B8, Canada
| | - Nicholas Erho
- Research and Development, GenomeDx Biosciences, Vancouver, BC V6B 1B8, Canada
| | - Paul Lloyd
- Department of Medicine, University of California at San Francisco, San Francisco, CA 94143-0410, USA; Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94143-0981, USA
| | | | - Ashley E Ross
- Brady Urological Institute, Johns Hopkins Medical Institute, Baltimore, MD 21287, USA
| | - Edward M Schaeffer
- Department of Urology, Northwestern University, Feinberg School of Medicine, 303 East Chicago Avenue, Tarry 16-703, Chicago, IL 60611-3008, USA
| | - Donald J Vander Griend
- Department of Surgery - Section of Urology, University of Chicago, Chicago, IL 60637, USA
| | - Stefan Knapp
- Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, UK; Institute for Pharmaceutical Chemistry, Goethe-University Frankfurt, Campus Riedberg, Max-von Laue Strasse 9, 60438 Frankfurt am Main, Germany
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA 98195, USA
| | - Felix Y Feng
- Department of Medicine, University of California at San Francisco, San Francisco, CA 94143-0410, USA; Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94143-0981, USA; Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Urology, University of Washington, Seattle, WA 98195, USA; Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Fahri Saatcioglu
- Department of Biosciences, University of Oslo, 0316 Oslo, Norway; Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0424 Oslo, Norway
| | - Karen E Knudsen
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Teuvo L J Tammela
- Prostate Cancer Research Center and Department of Urology, University of Tampere and Tampere University Hospital, 33014 Tampere, Finland
| | - Guido Sauter
- University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Thorsten Schlomm
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg 20095, Germany
| | - Matti Nykter
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Tampere University of Technology, 33520 Tampere, Finland
| | - Tapio Visakorpi
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Fimlab Laboratories, Tampere University Hospital, 33520 Tampere, Finland
| | - Ian G Mills
- Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, Forskningsparken, University of Oslo, 21 0349 Oslo, Norway; Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway; PCUK Movember Centre of Excellence, CCRCB, Queen's University, Belfast BT7 1NN, Northern Ireland, UK.
| |
Collapse
|
27
|
Epigenetic Signature: A New Player as Predictor of Clinically Significant Prostate Cancer (PCa) in Patients on Active Surveillance (AS). Int J Mol Sci 2017; 18:ijms18061146. [PMID: 28555004 PMCID: PMC5485970 DOI: 10.3390/ijms18061146] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 05/19/2017] [Accepted: 05/22/2017] [Indexed: 12/21/2022] Open
Abstract
Widespread prostate-specific antigen (PSA) testing notably increased the number of prostate cancer (PCa) diagnoses. However, about 30% of these patients have low-risk tumors that are not lethal and remain asymptomatic during their lifetime. Overtreatment of such patients may reduce quality of life and increase healthcare costs. Active surveillance (AS) has become an accepted alternative to immediate treatment in selected men with low-risk PCa. Despite much progress in recent years toward identifying the best candidates for AS in recent years, the greatest risk remains the possibility of misclassification of the cancer or missing a high-risk cancer. This is particularly worrisome in men with a life expectancy of greater than 10–15 years. The Prostate Cancer Research International Active Surveillance (PRIAS) study showed that, in addition to age and PSA at diagnosis, both PSA density (PSA-D) and the number of positive cores at diagnosis (two compared with one) are the strongest predictors for reclassification biopsy or switching to deferred treatment. However, there is still no consensus upon guidelines for placing patients on AS. Each institution has its own protocol for AS that is based on PRIAS criteria. Many different variables have been proposed as tools to enrol patients in AS: PSA-D, the percentage of freePSA, and the extent of cancer on biopsy (number of positive cores or percentage of core involvement). More recently, the Prostate Health Index (PHI), the 4 Kallikrein (4K) score, and other patient factors, such as age, race, and family history, have been investigated as tools able to predict clinically significant PCa. Recently, some reports suggested that epigenetic mapping differs significantly between cancer patients and healthy subjects. These findings indicated as future prospect the use of epigenetic markers to identify PCa patients with low-grade disease, who are likely candidates for AS. This review explores literature data about the potential of epigenetic markers as predictors of clinically significant disease.
Collapse
|
28
|
Pang J, Yang YW, Huang Y, Yang J, Zhang H, Chen R, Dong L, Huang Y, Wang D, Liu J, Li B. P110β Inhibition Reduces Histone H3K4 Di-Methylation in Prostate Cancer. Prostate 2017; 77:299-308. [PMID: 27800642 DOI: 10.1002/pros.23271] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 10/05/2016] [Indexed: 12/27/2022]
Abstract
INTRODUCTION AND AIMS Epigenetic alteration plays a major role in the development and progression of human cancers, including prostate cancer. Histones are the key factors in modulating gene accessibility to transcription factors and post-translational modification of the histone N-terminal tail including methylation is associated with either transcriptional activation (H3K4me2) or repression (H3K9me3). Furthermore, phosphoinositide 3-kinase (PI3 K) signaling and the androgen receptor (AR) are the key determinants in prostate cancer development and progression. We recently showed that prostate-targeted nano-micelles loaded with PI3 K/p110beta specific inhibitor TGX221 blocked prostate cancer growth in vitro and in vivo. Our objective of this study was to determine the role of PI3 K signaling in histone methylation in prostate cancer, with emphasis on histone H3K4 methylation. METHODS PI3 K non-specific inhibitor LY294002 and p110beta-specific inhibitor TGX221 were used to block PI3 K/p110beta signaling. The global levels of H3K4 and H3K9 methylation in prostate cancer cells and tissue specimens were evaluated by Western blot assay and immunohistochemical staining. A synthetic androgen R1881 was used to stimulate AR activity in prostate cancer cells. A castration-resistant prostate cancer (CRPC) specific human tissue microarray (TMA) was used to assess the global levels of H3K4me2 methylation by immunostaining approach. RESULTS Our data revealed that H3K4me2 levels were significantly elevated after androgen stimulation. With RNA silencing and pharmacology approaches, we further defined that inhibition of PI3 K/p110beta activity through gene-specific knocking down and small chemical inhibitor TGX221 abolished androgen-stimulated H3K4me2 methylation. Consistently, prostate cancer-targeted delivery of TGX221 in vivo dramatically reduced the global levels of H3K4me2 as assessed by immunohistochemical staining on tissue section of mouse xenografts from CRPC cell lines 22RV1 and C4-2. Finally, immunostaining data revealed a strong H3K4me2 immunosignal in CRPC tissues compared to primary tumors and benign prostate tissues. CONCLUSIONS Taken together, our results suggest that PI3 K/p110beta-dependent signaling is involved in androgen-stimulated H3K4me2 methylation in prostate cancer, which might be used as a novel biomarker for disease prognosis and targeted therapy. Prostate 77:299-308, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jun Pang
- Department of Urology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yue-Wu Yang
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yiling Huang
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
- Department of Pathology, China Three Gorges University School of Medicine, Yichang, China
| | - Jun Yang
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
- Department of Urology, Tongji Hospital, Huanzhong University of Science and Technology, Wuhan, China
| | - Hao Zhang
- Department of Urology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ruibao Chen
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
- Department of Urology, Tongji Hospital, Huanzhong University of Science and Technology, Wuhan, China
| | - Liang Dong
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Yan Huang
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Dongying Wang
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Jihong Liu
- Department of Urology, Tongji Hospital, Huanzhong University of Science and Technology, Wuhan, China
| | - Benyi Li
- Department of Urology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
- Department of Pathology, China Three Gorges University School of Medicine, Yichang, China
| |
Collapse
|
29
|
White-Al Habeeb NMA, Garcia J, Fleshner N, Bapat B. Metformin Elicits Antitumor Effects and Downregulates the Histone Methyltransferase Multiple Myeloma SET Domain (MMSET) in Prostate Cancer Cells. Prostate 2016; 76:1507-1518. [PMID: 27404348 DOI: 10.1002/pros.23235] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 06/29/2016] [Indexed: 11/06/2022]
Abstract
BACKGROUND This study explored the biological effects of metformin on prostate cancer (PCa) cells and determined molecular pathways and epigenetic regulators implicated in its mechanism of action. METHODS We performed mRNA expression profiling in 22Rv1 cells following 2.5 mM and 5 mM metformin treatment. Genes significantly modified by metformin treatment were ranked based on altered expression, involvement with cancer-related processes, and reported dysregulation in PCa. The effects of the top ranked gene, MMSET, on the proliferative and invasive capabilities of PCa cells were investigated via siRNA knockdown alone and also combined with metformin treatment. RESULTS Metformin treatment decreased cell growth of PCa cell line 22Rv1 and stalled cells at the G1/S checkpoint in a time- and dose-dependent manner, resulting in increased cells in G1 (P < 0.05) and decreased cells in S (P < 0.05) phase. Metformin activated the AMPK/mTOR signaling pathway as shown by increased p-AMPK and decreased p-p70S6K. mRNA expression profiling following metformin treatment identified significant changes in 136 chromatin-modifying genes. The top ranked gene, multiple myeloma SET domain (MMSET) showed increased expression in PCa cell lines (22Rv1 and DU145) when compared to the benign prostate epithelium-derived cell-line RWPE-1, and its expression was decreased upon metformin treatment. siRNA-mediated knockdown of MMSET showed decreased cellular migration and invasion in DU-145 cells. MMSET knockdown in combination with metformin treatment resulted in further reduction in the capacity of PCa cells to migrate and invade. CONCLUSIONS These data suggest MMSET may play a role in the inhibitory effect of metformin on PCa and could serve as a potential novel therapeutic target for PCa. Prostate 76:1507-1518, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nicole M A White-Al Habeeb
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Julia Garcia
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Neil Fleshner
- Ontario Cancer Institute, Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
| | - Bharati Bapat
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
- Department of Pathology, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
30
|
Fiano V, Zugna D, Grasso C, Trevisan M, Delsedime L, Molinaro L, Gillio-Tos A, Merletti F, Richiardi L. LINE-1 methylation status in prostate cancer and non-neoplastic tissue adjacent to tumor in association with mortality. Epigenetics 2016; 12:11-18. [PMID: 27892790 DOI: 10.1080/15592294.2016.1261786] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Aberrant DNA methylation seems to be associated with prostate cancer behavior. We investigated LINE-1 methylation in prostate cancer and non-neoplastic tissue adjacent to tumor (NTAT) in association with mortality from prostate cancer. We selected 157 prostate cancer patients with available NTAT from 2 cohorts of patients diagnosed between 1982-1988 and 1993-1996, followed up until 2010. An association between LINE-1 hypomethylation and prostate cancer mortality in tumor was suggested [hazard ratio per 5% decrease in LINE-1 methylation levels: 1.40, 95% confidence interval (CI): 0.95-2.01]. After stratification of the patients for Gleason score, the association was present only for those with a Gleason score of at least 8. Among these, low (<75%) vs. high (>80%) LINE-1 methylation was associated with a hazard ratio of 4.68 (95% CI: 1.03-21.34). LINE-1 methylation in the NTAT was not associated with prostate cancer mortality. Our results are consistent with the hypothesis that tumor tissue global hypomethylation may be a late event in prostate cancerogenesis and is associated with tumor progression.
Collapse
Affiliation(s)
- Valentina Fiano
- a Cancer Epidemiology Unit-CERMS , Department of Medical Sciences , University of Turin and CPO-Piemonte , Turin , Italy
| | - Daniela Zugna
- a Cancer Epidemiology Unit-CERMS , Department of Medical Sciences , University of Turin and CPO-Piemonte , Turin , Italy
| | - Chiara Grasso
- a Cancer Epidemiology Unit-CERMS , Department of Medical Sciences , University of Turin and CPO-Piemonte , Turin , Italy
| | - Morena Trevisan
- a Cancer Epidemiology Unit-CERMS , Department of Medical Sciences , University of Turin and CPO-Piemonte , Turin , Italy
| | - Luisa Delsedime
- b Division of Pathology, A.O. Città della Salute e della Scienza Hospital , Turin , Italy
| | - Luca Molinaro
- b Division of Pathology, A.O. Città della Salute e della Scienza Hospital , Turin , Italy
| | - Anna Gillio-Tos
- a Cancer Epidemiology Unit-CERMS , Department of Medical Sciences , University of Turin and CPO-Piemonte , Turin , Italy
| | - Franco Merletti
- a Cancer Epidemiology Unit-CERMS , Department of Medical Sciences , University of Turin and CPO-Piemonte , Turin , Italy
| | - Lorenzo Richiardi
- a Cancer Epidemiology Unit-CERMS , Department of Medical Sciences , University of Turin and CPO-Piemonte , Turin , Italy
| |
Collapse
|
31
|
Lee J, Han JH, Jang A, Kim JW, Hong SA, Myung SC. DNA Methylation-Mediated Downregulation of DEFB1 in Prostate Cancer Cells. PLoS One 2016; 11:e0166664. [PMID: 27835705 PMCID: PMC5105953 DOI: 10.1371/journal.pone.0166664] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/01/2016] [Indexed: 12/18/2022] Open
Abstract
Epigenetic aberrations play crucial roles in prostate cancer (PCa) development and progression. The DEFB1 gene, which encodes human ß-defensin-1 (HBD-1), contributes to innate immune responses and functions as a potential tumor suppressor in urological cancers. We investigated whether differential DNA methylation at the low CpG-content promoter (LCP) of DEFB1 was associated with transcriptional regulation of DEFB1 in PCa cells. To identify distinct CpG loci within the DEFB1 LCP related to the epigenetic regulation of DEFB1, we performed an in vitro methylated reporter assay followed by bisulfite sequencing of the DEFB1 promoter fragment. The methylation status of two adjacent CpG loci in the DEFB1 LCP was found to be important for DEFB1 expression in PCa cells. Paired epithelial specimens of PCa patients (n = 60), which were distinguished as non-tumor and tumor tissues by microdissection, were analyzed by bisulfite pyrosequencing of site-specific CpG dinucleotide units in the DEFB1 LCP. CpG methylation frequencies in the DEFB1 LCP were significantly higher in malignant tissues than in adjacent benign tissues across almost all PCa patients. These results suggested that methylation status of each CpG site in the DEFB1 promoter could mediate downregulation of DEFB1 in PCa cells.
Collapse
Affiliation(s)
- Jaehyouk Lee
- Department of Urology, Chung-Ang University College of Medicine, Seoul 06974, Republic of Korea
- Advanced Urogenital Diseases Research Center, Chung-Ang University College of Medicine, Seoul 06974, Republic of Korea
- Bio-Integration Research Center for Nutra-Pharmaceutical Epigenetics, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Jun Hyun Han
- Department of Urology, Hallym University Dongtan Sacred Heart Hospital, Hwaseong-si 18450, Republic of Korea
| | - Ara Jang
- Department of Urology, Chung-Ang University College of Medicine, Seoul 06974, Republic of Korea
- Advanced Urogenital Diseases Research Center, Chung-Ang University College of Medicine, Seoul 06974, Republic of Korea
- Bio-Integration Research Center for Nutra-Pharmaceutical Epigenetics, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Jin Wook Kim
- Bio-Integration Research Center for Nutra-Pharmaceutical Epigenetics, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Soon Auck Hong
- Department of Pathology, Soonchunhyang University College of Medicine, Cheonan 31151, Republic of Korea
| | - Soon Chul Myung
- Department of Urology, Chung-Ang University College of Medicine, Seoul 06974, Republic of Korea
- Advanced Urogenital Diseases Research Center, Chung-Ang University College of Medicine, Seoul 06974, Republic of Korea
- Bio-Integration Research Center for Nutra-Pharmaceutical Epigenetics, Chung-Ang University, Seoul 06974, Republic of Korea
- * E-mail:
| |
Collapse
|
32
|
Boyanapalli SSS, Li W, Fuentes F, Guo Y, Ramirez CN, Gonzalez XP, Pung D, Kong ANT. Epigenetic reactivation of RASSF1A by phenethyl isothiocyanate (PEITC) and promotion of apoptosis in LNCaP cells. Pharmacol Res 2016; 114:175-184. [PMID: 27818231 DOI: 10.1016/j.phrs.2016.10.021] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 10/21/2016] [Accepted: 10/24/2016] [Indexed: 12/23/2022]
Abstract
Epigenetic silencing of tumor suppressor genes is a phenomenon frequently observed in multiple cancers. Ras-association domain family 1 isoform A (RASSF1A) is a well-characterized tumor suppressor that belongs to the Ras-association domain family. Several studies have demonstrated that hypermethylation of the RASSF1A promoter is frequently observed in lung, prostate, and breast cancers. Phenethyl isothiocyanate (PEITC), a phytochemical abundant in cruciferous vegetables, possesses chemopreventive activities; however, its potential involvement in epigenetic mechanisms remains elusive. The present study aimed to examine the role of PEITC in the epigenetic reactivation of RASSF1A and the induction of apoptosis in LNCaP cells. LNCaP cells were treated for 5days with 0.01% DMSO, 2.5 or 5μM PETIC or 2.5μM azadeoxycytidine (5-Aza) with 0.5μM trichostatin A (TSA). We evaluated the effects of these treatments on CpG demethylation using methylation-specific polymerase chain reaction (MSP) and bisulfite genomic sequencing (BGS). CpG demethylation was significantly enhanced in cells treated with 5μM PEITC and 5-Aza+TSA; therefore, the latter treatment was used as a positive control in subsequent experiments. The decrease in RASSF1A promoter methylation correlated with an increase in expression of the RASSF1A gene in a dose-dependent manner. To confirm that promoter demethylation was mediated by DNA methyltransferases (DNMTs), we analyzed the expression levels of DNMTs and histone deacetylases (HDACs) at the gene and protein levels. PEITC reduced DNMT1, 3A and 3B protein levels in a dose-dependent manner, and 5μM PEITC significantly reduced DNMT3A and 3B protein levels. HDAC1, 2, 4 and 6 protein expression was also inhibited by 5μM PEITC. The combination of 5-Aza and TSA, a DNMT inhibitor and a HDAC inhibitor, respectively, was used as a positive control as this treatment significantly inhibited both HDACs and DNMTs. The function of RASSF1A reactivation in promoting apoptosis and inducing G2/M cell cycle arrest was analyzed using flow-cytometry analysis with Annexin V and propidium iodide (PI). Growth inhibition effect on LNCaP cells were investigated by colony formation assay. In addition, we analyzed p21, caspase-3 and 7, Bax, and Cyclin B1 protein levels. Flow-cytometry analysis of cells stained with PI alone demonstrated that 5μM PEITC promotes early apoptosis and G2/M cell cycle arrest. Flow cytometry analysis of cells stained with Annexin V and PI also demonstrated an increased proportion of cells in early apoptosis in cells treated with 5μM PEITC or 5-Aza with TSA. PEITC and efficiently inhibit colony numbers and total area. In addition, 5μM PEITC significantly enhanced p21, caspase-3, 7 and Bax levels and reduced Cyclin B1 expression compared with the control group. Collectively, the results of our study suggest that PEITC induces apoptosis in LNCaP cells potentially by reactivating RASSF1A via epigenetic mechanisms.
Collapse
Affiliation(s)
- Sarandeep S S Boyanapalli
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest-Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road Piscataway, NJ, 08854, United States
| | - Wenji Li
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest-Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road Piscataway, NJ, 08854, United States
| | - Francisco Fuentes
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest-Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road Piscataway, NJ, 08854, United States
| | - Yue Guo
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest-Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road Piscataway, NJ, 08854, United States
| | - Christina N Ramirez
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest-Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road Piscataway, NJ, 08854, United States
| | - Ximena-Parades Gonzalez
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest-Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road Piscataway, NJ, 08854, United States
| | - Douglas Pung
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest-Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road Piscataway, NJ, 08854, United States
| | - Ah-Ng Tony Kong
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest-Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road Piscataway, NJ, 08854, United States.
| |
Collapse
|
33
|
A new transgenic mouse model for conditional overexpression of the Polycomb Group protein EZH2. Transgenic Res 2016; 26:187-196. [DOI: 10.1007/s11248-016-9993-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/25/2016] [Indexed: 01/12/2023]
|
34
|
Graça I, Pereira-Silva E, Henrique R, Packham G, Crabb SJ, Jerónimo C. Epigenetic modulators as therapeutic targets in prostate cancer. Clin Epigenetics 2016; 8:98. [PMID: 27651838 PMCID: PMC5025578 DOI: 10.1186/s13148-016-0264-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/07/2016] [Indexed: 01/24/2023] Open
Abstract
Prostate cancer is one of the most common non-cutaneous malignancies among men worldwide. Epigenetic aberrations, including changes in DNA methylation patterns and/or histone modifications, are key drivers of prostate carcinogenesis. These epigenetic defects might be due to deregulated function and/or expression of the epigenetic machinery, affecting the expression of several important genes. Remarkably, epigenetic modifications are reversible and numerous compounds that target the epigenetic enzymes and regulatory proteins were reported to be effective in cancer growth control. In fact, some of these drugs are already being tested in clinical trials. This review discusses the most important epigenetic alterations in prostate cancer, highlighting the role of epigenetic modulating compounds in pre-clinical and clinical trials as potential therapeutic agents for prostate cancer management.
Collapse
Affiliation(s)
- Inês Graça
- Cancer Biology and Epigenetics Group-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Research Center-LAB 3, F Bdg, 1st floor, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal ; School of Allied Health Sciences (ESTSP), Polytechnic of Porto, Porto, Portugal
| | - Eva Pereira-Silva
- Cancer Biology and Epigenetics Group-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Research Center-LAB 3, F Bdg, 1st floor, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Research Center-LAB 3, F Bdg, 1st floor, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal ; Department of Pathology, Portuguese Oncology Institute of Porto (IPO 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 (ICBAS-UP), Porto, Portugal
| | - Graham Packham
- Cancer Research UK Centre, Cancer Sciences, The Somers Cancer Research Building, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, S016 6YD UK
| | - Simon J Crabb
- Cancer Research UK Centre, Cancer Sciences, The Somers Cancer Research Building, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, S016 6YD UK
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Research Center-LAB 3, F Bdg, 1st floor, 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 (ICBAS-UP), Porto, Portugal
| |
Collapse
|
35
|
Corbin JM, Ruiz-Echevarría MJ. One-Carbon Metabolism in Prostate Cancer: The Role of Androgen Signaling. Int J Mol Sci 2016; 17:E1208. [PMID: 27472325 PMCID: PMC5000606 DOI: 10.3390/ijms17081208] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 07/16/2016] [Accepted: 07/18/2016] [Indexed: 01/06/2023] Open
Abstract
Cancer cell metabolism differs significantly from the metabolism of non-transformed cells. This altered metabolic reprogramming mediates changes in the uptake and use of nutrients that permit high rates of proliferation, growth, and survival. The androgen receptor (AR) plays an essential role in the establishment and progression of prostate cancer (PCa), and in the metabolic adaptation that takes place during this progression. In its role as a transcription factor, the AR directly affects the expression of several effectors and regulators of essential catabolic and biosynthetic pathways. Indirectly, as a modulator of the one-carbon metabolism, the AR can affect epigenetic processes, DNA metabolism, and redox balance, all of which are important factors in tumorigenesis. In this review, we focus on the role of AR-signaling on one-carbon metabolism in tumorigenesis. Clinical implications of one-carbon metabolism and AR-targeted therapies for PCa are discussed in this context.
Collapse
Affiliation(s)
- Joshua M Corbin
- Department of Pathology, Oklahoma University Health Sciences Center, Oklahoma City, OK 73104, USA.
| | - Maria J Ruiz-Echevarría
- Department of Pathology, Oklahoma University Health Sciences Center and Stephenson Cancer Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
36
|
Yen CY, Huang HW, Shu CW, Hou MF, Yuan SSF, Wang HR, Chang YT, Farooqi AA, Tang JY, Chang HW. DNA methylation, histone acetylation and methylation of epigenetic modifications as a therapeutic approach for cancers. Cancer Lett 2016; 373:185-92. [DOI: 10.1016/j.canlet.2016.01.036] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/23/2015] [Accepted: 01/18/2016] [Indexed: 02/09/2023]
|
37
|
Identification of the epigenetic reader CBX2 as a potential drug target in advanced prostate cancer. Clin Epigenetics 2016; 8:16. [PMID: 26877821 PMCID: PMC4751702 DOI: 10.1186/s13148-016-0182-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 02/04/2016] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND While localized prostate cancer (PCa) can be effectively cured, metastatic disease inevitably progresses to a lethal state called castration-resistant prostate cancer (CRPC). Emerging evidence suggests that aberrant epigenetic repression by the polycomb group (PcG) complexes fuels PCa progression, providing novel therapeutic opportunities. RESULTS In the search for potential epigenetic drivers of CRPC, we analyzed the molecular profile of PcG members in patient-derived xenografts and clinical samples. Overall, our results identify the PcG protein and methyl-lysine reader CBX2 as a potential therapeutic target in advanced PCa. We report that CBX2 was recurrently up-regulated in metastatic CRPC and that elevated CBX2 expression was correlated with poor clinical outcome in PCa cohorts. Furthermore, CBX2 depletion abrogated cell viability and induced caspase 3-mediated apoptosis in metastatic PCa cell lines. Mechanistically explaining this phenotype, microarray analysis in CBX2-depleted cells revealed that CBX2 controls the expression of many key regulators of cell proliferation and metastasis. CONCLUSIONS Taken together, this study provides the first evidence that CBX2 inhibition induces cancer cell death, positioning CBX2 as an attractive drug target in lethal CRPC.
Collapse
|
38
|
Elmer JJ, Christensen MD, Barua S, Lehrman J, Haynes KA, Rege K. The histone deacetylase inhibitor Entinostat enhances polymer-mediated transgene expression in cancer cell lines. Biotechnol Bioeng 2015; 113:1345-1356. [PMID: 26614912 DOI: 10.1002/bit.25898] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 10/26/2015] [Accepted: 11/25/2015] [Indexed: 12/31/2022]
Abstract
Eukaryotic cells maintain an immense amount of genetic information by tightly wrapping their DNA around positively charged histones. While this strategy allows human cells to maintain more than 25,000 genes, histone binding can also block gene expression. Consequently, cells express histone acetyl transferases (HATs) to acetylate histone lysines and release DNA for transcription. Conversely, histone deacetylases (HDACs) are employed for restoring the positive charge on the histones, thereby silencing gene expression by increasing histone-DNA binding. It has previously been shown that histones bind and silence viral DNA, while hyperacetylation of histones via HDAC inhibition restores viral gene expression. In this study, we demonstrate that treatment with Entinostat, an HDAC inhibitor, enhances transgene (luciferase) expression by up to 25-fold in human prostate and murine bladder cancer cell lines when used with cationic polymers for plasmid DNA delivery. Entinostat treatment altered cell cycle progression, resulting in a significant increase in the fraction of cells present in the G0/G1 phase at low micromolar concentrations. While this moderate G0/G1 arrest disappeared at higher concentrations, a modest increase in the fraction of apoptotic cells and a decrease in cell proliferation were observed, consistent with the known anticancer effects of the drug. DNase accessibility studies revealed no significant change in plasmid transcriptional availability with Entinostat treatment. However, quantitative PCR studies indicated that Entinostat treatment, at the optimal dose for enhancing transgene expression, led to an increase in the amount of plasmid present in the nucleus in two cancer cell lines. Taken together, our results show that Entinostat enhances polymer- mediated transgene expression and can be useful in applications related to transient protein expression in mammalian cells. Biotechnol. Bioeng. 2016;113: 1345-1356. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jacob J Elmer
- Chemical Engineering, Arizona State University, Tempe, Arizona 85287
| | | | - Sutapa Barua
- Chemical Engineering, Arizona State University, Tempe, Arizona 85287
| | - Jennifer Lehrman
- Harrington Biomedical Engineering, Arizona State University, Tempe, Arizona
| | - Karmella A Haynes
- Harrington Biomedical Engineering, Arizona State University, Tempe, Arizona
| | - Kaushal Rege
- Chemical Engineering, Arizona State University, Tempe, Arizona 85287
| |
Collapse
|
39
|
Kaushik D, Vashistha V, Isharwal S, Sediqe SA, Lin MF. Histone deacetylase inhibitors in castration-resistant prostate cancer: molecular mechanism of action and recent clinical trials. Ther Adv Urol 2015; 7:388-95. [PMID: 26622323 DOI: 10.1177/1756287215597637] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Historically, androgen-deprivation therapy has been the cornerstone for treatment of metastatic prostate cancer. Unfortunately, nearly majority patients with prostate cancer transition to the refractory state of castration-resistant prostate cancer (CRPC). Newer therapeutic agents are needed for treating these CRPC patients that are unresponsive to androgen deprivation and/or chemotherapy. The histone deacetylase (HDAC) family of enzymes limits the expression of genomic regions by improving binding between histones and the DNA backbone. Modulating the role of HDAC enzymes can alter the cell's regulation of proto-oncogenes and tumor suppressor genes, thereby regulating potential neoplastic proliferation. As a result, histone deacetylase inhibitors (HDACi) are now being evaluated for CRPC or chemotherapy-resistant prostate cancer due to their effects on the expression of the androgen receptor gene. In this paper, we review the molecular mechanism and functional target molecules of different HDACi as applicable to CRPC as well as describe recent and current clinical trials involving HDACi in prostate cancer. To date, four HDAC classes comprising 18 isoenzymes have been identified. Recent clinical trials of vorinostat, romidepsin, and panobinostat have provided cautious optimism towards improved outcomes using these novel therapeutic agents for CPRC patients. Nevertheless, no phase III trial has been conducted to cement one of these drugs as an adjunct to androgen-deprivation therapy. Consequently, further investigation is necessary to delineate the benefits and drawbacks of these medications.
Collapse
Affiliation(s)
- Dharam Kaushik
- Department of Urology, University of Texas Health Science Center and Cancer Therapy and Research Center, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, USA
| | - Vishal Vashistha
- Department of Internal Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Sudhir Isharwal
- Section of Urology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Soud A Sediqe
- Department of Internal Medicine, MetroHealth Medical Center, Cleveland, OH, USA
| | - Ming-Fong Lin
- Section of Urology, and Department of Biochemistry, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
40
|
Sharma V, Verma V, Lal N, Yadav SK, Sarkar S, Mandalapu D, Porwal K, Rawat T, Maikhuri JP, Rajender S, Sharma VL, Gupta G. Disulfiram and its novel derivative sensitize prostate cancer cells to the growth regulatory mechanisms of the cell by re-expressing the epigenetically repressed tumor suppressor-estrogen receptor β. Mol Carcinog 2015; 55:1843-1857. [PMID: 26599461 DOI: 10.1002/mc.22433] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 10/20/2015] [Accepted: 11/03/2015] [Indexed: 11/09/2022]
Abstract
Estrogen Receptor-β (ER-β), a tumor-suppressor in prostate cancer, is epigenetically repressed by hypermethylation of its promoter. DNA-methyltransferases (DNMTs), which catalyze the transfer of methyl-groups to CpG islands of gene promoters, are overactive in cancers and can be inhibited by DNMT-inhibitors to re-express the tumor suppressors. The FDA-approved nucleoside DNMT-inhibitors like 5-Azacytidine and 5-Aza-deoxycytidine carry notable concerns due to their off-target toxicity, therefore non-nucleoside DNMT inhibitors are desirable for prolonged epigenetic therapy. Disulfiram (DSF), an antabuse drug, inhibits DNMT and prevents proliferation of cells in prostate and other cancers, plausibly through the re-expression of tumor suppressors like ER-β. To increase the DNMT-inhibitory activity of DSF, its chemical scaffold was optimized and compound-339 was discovered as a doubly potent DSF-derivative with similar off-target toxicity. It potently and selectively inhibited cell proliferation of prostate cancer (PC3/DU145) cells in comparison to normal (non-cancer) cells by promoting cell-cycle arrest and apoptosis, accompanied with inhibition of total DNMT activity, and re-expression of ER-β (mRNA/protein). Bisulfite-sequencing of ER-β promoter revealed that compound-339 demethylated CpG sites more efficaciously than DSF, restoring near-normal methylation status of ER-β promoter. Compound-339 docked on to the MTase domain of DNMT1 with half the energy of DSF. In xenograft mice-model, the tumor volume regressed by 24% and 50% after treatment with DSF and compound-339, respectively, with increase in ER-β expression. Apparently both compounds inhibit prostate cancer cell proliferation by re-expressing the epigenetically repressed tumor-suppressor ER-β through inhibition of DNMT activity. Compound-339 presents a new lead for further study as an anti-prostate cancer agent. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Vikas Sharma
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Vikas Verma
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Nand Lal
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, India
| | - Santosh K Yadav
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Saumya Sarkar
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Dhanaraju Mandalapu
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, India
| | - Konica Porwal
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Tara Rawat
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, India
| | - J P Maikhuri
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Singh Rajender
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - V L Sharma
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, India
| | - Gopal Gupta
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India.
| |
Collapse
|
41
|
Labbé DP, Zadra G, Ebot EM, Mucci LA, Kantoff PW, Loda M, Brown M. Role of diet in prostate cancer: the epigenetic link. Oncogene 2015; 34:4683-91. [PMID: 25531313 PMCID: PMC4476943 DOI: 10.1038/onc.2014.422] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 10/28/2014] [Accepted: 11/03/2014] [Indexed: 12/12/2022]
Abstract
Diet is hypothesized to be a critical environmentally related risk factor for prostate cancer (PCa) development, and specific diets and dietary components can also affect PCa progression; however, the mechanisms underlying these associations remain elusive. As for a maturing organism, PCa's epigenome is plastic and evolves from the pre-neoplastic to the metastatic stage. In particular, epigenetic remodeling relies on substrates or cofactors obtained from the diet. Here we review the evidence that bridges dietary modulation to alterations in the prostate epigenome. We propose that such diet-related effects offer a mechanistic link between the impact of different diets and the course of PCa development and progression.
Collapse
Affiliation(s)
- D P Labbé
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - G Zadra
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - E M Ebot
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
| | - L A Mucci
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
- Channing Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - P W Kantoff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - M Loda
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - M Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
42
|
DNA Methylation-Guided Prediction of Clinical Failure in High-Risk Prostate Cancer. PLoS One 2015; 10:e0130651. [PMID: 26086362 PMCID: PMC4472347 DOI: 10.1371/journal.pone.0130651] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 05/25/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Prostate cancer (PCa) is a very heterogeneous disease with respect to clinical outcome. This study explored differential DNA methylation in a priori selected genes to diagnose PCa and predict clinical failure (CF) in high-risk patients. METHODS A quantitative multiplex, methylation-specific PCR assay was developed to assess promoter methylation of the APC, CCND2, GSTP1, PTGS2 and RARB genes in formalin-fixed, paraffin-embedded tissue samples from 42 patients with benign prostatic hyperplasia and radical prostatectomy specimens of patients with high-risk PCa, encompassing training and validation cohorts of 147 and 71 patients, respectively. Log-rank tests, univariate and multivariate Cox models were used to investigate the prognostic value of the DNA methylation. RESULTS Hypermethylation of APC, CCND2, GSTP1, PTGS2 and RARB was highly cancer-specific. However, only GSTP1 methylation was significantly associated with CF in both independent high-risk PCa cohorts. Importantly, trichotomization into low, moderate and high GSTP1 methylation level subgroups was highly predictive for CF. Patients with either a low or high GSTP1 methylation level, as compared to the moderate methylation groups, were at a higher risk for CF in both the training (Hazard ratio [HR], 3.65; 95% CI, 1.65 to 8.07) and validation sets (HR, 4.27; 95% CI, 1.03 to 17.72) as well as in the combined cohort (HR, 2.74; 95% CI, 1.42 to 5.27) in multivariate analysis. CONCLUSIONS Classification of primary high-risk tumors into three subtypes based on DNA methylation can be combined with clinico-pathological parameters for a more informative risk-stratification of these PCa patients.
Collapse
|
43
|
Atwell LL, Beaver LM, Shannon J, Williams DE, Dashwood RH, Ho E. Epigenetic Regulation by Sulforaphane: Opportunities for Breast and Prostate Cancer Chemoprevention. ACTA ACUST UNITED AC 2015; 1:102-111. [PMID: 26042194 DOI: 10.1007/s40495-014-0002-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Sulforaphane (SFN) is a phytochemical derived from cruciferous vegetables that has multiple molecular targets and anti-cancer properties. Researchers have demonstrated several chemopreventive benefits of SFN consumption, such as reductions in tumor growth, increases in cancer cell apoptosis, and disruption of signaling within tumor microenvironments both in vitro and in vivo. Emerging evidence indicates that SFN exerts several of its chemopreventive effects by altering epigenetic mechanisms. This review summarizes evidence of the impact of SFN on epigenetic events and how they relate to the chemopreventive effects of SFN observed in preclinical and clinical studies of breast and prostate cancers. Specific areas of focus include the role of SFN in the regulation of cell cycle, apoptosis, inflammation, antioxidant defense, and cancer cell signaling and their relationships to epigenetic mechanisms. Finally, remaining challenges and research needs for translating mechanistic work with SFN into human studies and clinical intervention trials are discussed.
Collapse
Affiliation(s)
- Lauren L Atwell
- 103 Milam Hall, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Laura M Beaver
- 103 Milam Hall, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA ; 307 Linus Pauling Science Center, Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | - Jackilen Shannon
- 3181 SW Sam Jackson Park Road, Mail Code CB L606, Department of Public Health and Preventive Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - David E Williams
- 307 Linus Pauling Science Center, Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA ; 1007 Agriculture & Life Sciences Building, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Roderick H Dashwood
- 2121 West Holcombe Boulevard, Center for Epigenetics & Disease Prevention, Houston, TX 77030
| | - Emily Ho
- 103 Milam Hall, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA ; 307 Linus Pauling Science Center, Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA ; 212 Milam Hall, Moore Family Center for Whole Grain Foods, Nutrition and Preventive Health, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
44
|
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
|
45
|
Abstract
The introduction of novel technologies that can be applied to the investigation of the molecular underpinnings of human cancer has allowed for new insights into the mechanisms associated with tumor development and progression. They have also advanced the diagnosis, prognosis and treatment of cancer. These technologies include microarray and other analysis methods for the generation of large-scale gene expression data on both mRNA and miRNA, next-generation DNA sequencing technologies utilizing a number of platforms to perform whole genome, whole exome, or targeted DNA sequencing to determine somatic mutational differences and gene rearrangements, and a variety of proteomic analysis platforms including liquid chromatography/mass spectrometry (LC/MS) analysis to survey alterations in protein profiles in tumors. One other important advancement has been our current ability to survey the methylome of human tumors in a comprehensive fashion through the use of sequence-based and array-based methylation analysis (Bock et al., Nat Biotechnol 28:1106-1114, 2010; Harris et al., Nat Biotechnol 28:1097-1105, 2010). The focus of this chapter is to present and discuss the evidence for key genes involved in prostate tumor development, progression, or resistance to therapy that are regulated by methylation-induced silencing.
Collapse
Affiliation(s)
- Tawnya C McKee
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, Diagnostic Biomarkers and Technology Branch, National Cancer Institute, Bethesda, MD, 20892-7430, USA
| | | |
Collapse
|
46
|
Functional Epigenetic Analysis of Prostate Carcinoma: A Role for Seryl-tRNA Synthetase? J Biomark 2014; 2014:362164. [PMID: 26317032 PMCID: PMC4437382 DOI: 10.1155/2014/362164] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 02/21/2014] [Indexed: 01/10/2023] Open
Abstract
Transcriptional silencing, as a result of aberrant promoter hypermethylation, is a common mechanism through which genes in cancer cells become inactive. Functional epigenetic screens using demethylating agents to reexpress transcriptional silenced genes may identify such inactivated genes for needing further evaluation. We aimed to identify genes so far not known to be inactivated by promoter hypermethylation in prostate cancer. DU-145 and LNCaP cells were treated with the DNMT inhibitor zebularine. Expression changes of total RNA from treated and untreated cells were compared using an RNA expression microarray. Genes upregulated more than 2-fold were evaluated by RT-qPCR in 50 cases of paired normal and tumor tissues of prostate cancer patients. SARS was found to be downregulated in prostate cancer in 42/50 cases (84%). In addition, GADD45A and SPRY4 showed a remarkable diminished expression (88% and 74%, resp.). The gold standard for promoter hypermethylation-inactivated genes in prostate cancer (GSTP1) was repressed in 90% of our patient samples. ROC analyses reported statistically significant AUC curves in SARS, GADD45A, and GSTP1 and positive Spearman correlations were found between these genes. SARS was discovered to be a novel gene that is repressed in prostate cancer and could therefore be recommended for its involvement in prostate carcinogenesis.
Collapse
|
47
|
Batra JS, Girdhani S, Hlatky L. A Quest to Identify Prostate Cancer Circulating Biomarkers with a Bench-to-Bedside Potential. J Biomark 2014; 2014:321680. [PMID: 26317031 PMCID: PMC4437363 DOI: 10.1155/2014/321680] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 01/07/2014] [Accepted: 01/10/2014] [Indexed: 02/01/2023] Open
Abstract
Prostate cancer (PCA) is a major health concern in current times. Ever since prostate specific antigen (PSA) was introduced in clinical practice almost three decades ago, the diagnosis and management of PCA have been revolutionized. With time, concerns arose as to the inherent shortcomings of this biomarker and alternatives were actively sought. Over the past decade new PCA biomarkers have been identified in tissue, blood, urine, and other body fluids that offer improved specificity and supplement our knowledge of disease progression. This review focuses on superiority of circulating biomarkers over tissue biomarkers due to the advantages of being more readily accessible, minimally invasive (blood) or noninvasive (urine), accessible for sampling on regular intervals, and easily utilized for follow-up after surgery or other treatment modalities. Some of the circulating biomarkers like PCA3, IL-6, and TMPRSS2-ERG are now detectable by commercially available kits while others like microRNAs (miR-21, -221, -141) and exosomes hold potential to become available as multiplexed assays. In this paper, we will review some of these potential candidate circulating biomarkers that either individually or in combination, once validated with large-scale trials, may eventually get utilized clinically for improved diagnosis, risk stratification, and treatment.
Collapse
Affiliation(s)
- Jaspreet Singh Batra
- Center of Cancer Systems Biology, GeneSys Research Institute, Tufts University, School of Medicine, 736 Cambridge Street, SEMC-CBR112, Boston, MA 02135, USA
| | - Swati Girdhani
- Center of Cancer Systems Biology, GeneSys Research Institute, Tufts University, School of Medicine, 736 Cambridge Street, SEMC-CBR112, Boston, MA 02135, USA
| | - Lynn Hlatky
- Center of Cancer Systems Biology, GeneSys Research Institute, Tufts University, School of Medicine, 736 Cambridge Street, SEMC-CBR112, Boston, MA 02135, USA
| |
Collapse
|
48
|
Surana R, Sikka S, Cai W, Shin EM, Warrier SR, Tan HJG, Arfuso F, Fox SA, Dharmarajan AM, Kumar AP. Secreted frizzled related proteins: Implications in cancers. Biochim Biophys Acta Rev Cancer 2013; 1845:53-65. [PMID: 24316024 DOI: 10.1016/j.bbcan.2013.11.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Revised: 11/26/2013] [Accepted: 11/30/2013] [Indexed: 12/31/2022]
Abstract
The Wnt (wingless-type) signaling pathway plays an important role in embryonic development, tissue homeostasis, and tumor progression becaluse of its effect on cell proliferation, migration, and differentiation. Secreted frizzled-related proteins (SFRPs) are extracellular inhibitors of Wnt signaling that act by binding directly to Wnt ligands or to Frizzled receptors. In recent years, aberrant expression of SFRPs has been reported to be associated with numerous cancers. As gene expression of SFRP members is often lost through promoter hypermethylation, inhibition of methylation through the use of epigenetic modifying agents could renew the expression of SFRP members and further antagonize deleterious Wnt signaling. Several reports have described epigenetic silencing of these Wnt signaling antagonists in various human cancers, suggesting their possible role as tumor suppressors. SFRP family members thus come across as potential tools in combating Wnt-driven tumorigenesis. However, little is known about SFRP family members and their role in different cancers. This review comprehensively covers all the available information on the role of SFRP molecules in various human cancers.
Collapse
Affiliation(s)
- Rohit Surana
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sakshi Sikka
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wanpei Cai
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Eun Myoung Shin
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Sudha R Warrier
- Manipal Institute of Regenerative Medicine, Manipal University, Bangalore, India
| | - Hong Jie Gabriel Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Frank Arfuso
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia, Australia; School of Biomedical Sciences, Faculty of Health Sciences, Curtin University, Perth, 6845 Western Australia, Australia
| | - Simon A Fox
- Molecular Pharmacology Laboratory, School of Pharmacy, Western Australian Biomedical Research Institute & Curtin Health Innovation Research Institute, Curtin University, Bentley, Australia
| | - Arun M Dharmarajan
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia, Australia; School of Biomedical Sciences, Faculty of Health Sciences, Curtin University, Perth, 6845 Western Australia, Australia.
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; School of Biomedical Sciences, Faculty of Health Sciences, Curtin University, Perth, 6845 Western Australia, Australia; Department of Biological Sciences, University of North Texas, Denton, TX 76203-5017, USA.
| |
Collapse
|
49
|
Chung PH, Gayed BA, Thoreson GR, Raj GV. Emerging drugs for prostate cancer. Expert Opin Emerg Drugs 2013; 18:533-50. [DOI: 10.1517/14728214.2013.864635] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
50
|
Gryder BE, Akbashev MJ, Rood MK, Raftery ED, Meyers WM, Dillard P, Khan S, Oyelere AK. Selectively targeting prostate cancer with antiandrogen equipped histone deacetylase inhibitors. ACS Chem Biol 2013; 8:2550-60. [PMID: 24004176 DOI: 10.1021/cb400542w] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Diverse cellular processes relevant to cancer progression are regulated by the acetylation status of proteins. Among such processes is chromatin remodeling via histone proteins, controlled by opposing histone deacetylase (HDAC) and histone acetyltransferase (HAT) enzymes. Histone deacetylase inhibitors (HDACi) show great promise in preclinical cancer models, but clinical trials treating solid tumors have failed to improve patient survival. This is due in part to an inability of HDACi to effectively accumulate in cancerous cells. To address this problem we designed HDACi with secondary pharmacophores to facilitate selective accumulation in malignant cells. We present the first example of HDACi compounds targeted to prostate tumors by equipping them with the additional ability to bind the androgen receptor (AR) with nonsteroidal antiandrogen moieties. Leads among these new dual-acting molecules bind to the AR and halt AR transcriptional activity at lower concentrations than clinical antiandrogens. They inhibit key isoforms of HDAC with low nanomolar potency. Fluorescent microscopy reveals varying degrees of AR nuclear localization in response to these compounds that correlates with their HDAC activity. These biological properties translate into potent anticancer activity against hormone-dependent (AR+) LNCaP and to a lesser extent against hormone-independent (AR-) DU145 prostate cancer, while having greatly reduced toxicity in noncancerous cells. This illustrates that engaging multiple biological targets with a single chemical probe can achieve both potent and cell-type-selective responses.
Collapse
Affiliation(s)
- Berkley E. Gryder
- Parker H. Petit Institute for Bioengineering & Biosciences, Department of Chemistry and Biochemistry, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, Georgia 30332-0230, United States
| | - Michelle J. Akbashev
- Parker H. Petit Institute for Bioengineering & Biosciences, Department of Chemistry and Biochemistry, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, Georgia 30332-0230, United States
| | - Michael K. Rood
- Parker H. Petit Institute for Bioengineering & Biosciences, Department of Chemistry and Biochemistry, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, Georgia 30332-0230, United States
| | - Eric D. Raftery
- Parker H. Petit Institute for Bioengineering & Biosciences, Department of Chemistry and Biochemistry, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, Georgia 30332-0230, United States
| | - Warren M. Meyers
- Department of Cellular
and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Paulette Dillard
- Center
for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, Georgia 30314, United States
| | - Shafiq Khan
- Center
for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, Georgia 30314, United States
| | - Adegboyega K. Oyelere
- Parker H. Petit Institute for Bioengineering & Biosciences, Department of Chemistry and Biochemistry, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, Georgia 30332-0230, United States
| |
Collapse
|