51
|
Al-Moghrabi N, Al-Showimi M, Al-Yousef N, Al-Shahrani B, Karakas B, Alghofaili L, Almubarak H, Madkhali S, Al Humaidan H. Methylation of BRCA1 and MGMT genes in white blood cells are transmitted from mothers to daughters. Clin Epigenetics 2018; 10:99. [PMID: 30049288 PMCID: PMC6062990 DOI: 10.1186/s13148-018-0529-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/08/2018] [Indexed: 12/31/2022] Open
Abstract
Background Constitutive methylation of tumor suppressor genes are associated with increased cancer risk. However, to date, the question of epimutational transmission of these genes remains unresolved. Here, we studied the potential transmission of BRCA1 and MGMT promoter methylations in mother-newborn pairs. Methods A total of 1014 female subjects (cancer-free women, n = 268; delivering women, n = 295; newborn females, n = 302; breast cancer patients, n = 67; ovarian cancer patients, n = 82) were screened for methylation status in white blood cells (WBC) using methylation-specific PCR and bisulfite pyrosequencing assays. In addition, BRCA1 gene expression levels were analyzed by quantitative real-time PCR. Results We found similar methylation frequencies in newborn and adults for both BRCA1 (9.9 and 9.3%) and MGMT (12.3 and 13.1%). Of the 290 mother-newborn pairs analyzed for promoter methylation, 20 mothers were found to be positive for BRCA1 and 29 for MGMT. Four mother-newborn pairs were positive for methylated BRCA1 (20%) and nine pairs were positive for methylated MGMT (31%). Intriguingly, the delivering women had 26% lower BRCA1 and MGMT methylation frequencies than those of the cancer-free female subjects. BRCA1 was downregulated in both cancer-free woman carriers and breast cancer patients but not in newborn carriers. There was a statistically significant association between the MGMT promoter methylation and late-onset breast cancers. Conclusions Our study demonstrates that BRCA1and MGMT epimutations are present from the early life of the carriers. We show the transmission of BRCA1 and MGMT epimutations from mother to daughter. Our data also point at the possible demethylation of BRCA1and MGMT during pregnancy.
Collapse
Affiliation(s)
- Nisreen Al-Moghrabi
- Head of Cancer Epigenetic Section, Molecular Oncology Department, King Faisal Specialist Hospital and Research Centre, PO BOX 3354, Riyadh, 11211, Kingdom of Saudi Arabia.
| | - Maram Al-Showimi
- Cancer Epigenetic section, Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, PO BOX 3354, Riyadh, 11211, Kingdom of Saudi Arabia
| | - Nujoud Al-Yousef
- Head of Cancer Epigenetic Section, Molecular Oncology Department, King Faisal Specialist Hospital and Research Centre, PO BOX 3354, Riyadh, 11211, Kingdom of Saudi Arabia
| | - Bushra Al-Shahrani
- Cancer Epigenetic section, Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, PO BOX 3354, Riyadh, 11211, Kingdom of Saudi Arabia
| | - Bedri Karakas
- Head of Cancer Epigenetic Section, Molecular Oncology Department, King Faisal Specialist Hospital and Research Centre, PO BOX 3354, Riyadh, 11211, Kingdom of Saudi Arabia
| | - Lamyaa Alghofaili
- Al Faisal University College of Medicine, PO BOX 50927, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Hannah Almubarak
- Head of Cancer Epigenetic Section, Molecular Oncology Department, King Faisal Specialist Hospital and Research Centre, PO BOX 3354, Riyadh, 11211, Kingdom of Saudi Arabia
| | - Safia Madkhali
- King Saud bin Abdulaziz University for Health Sciences, PO BOX 22490, Riyadh, 3130, Kingdom of Saudi Arabia
| | - Hind Al Humaidan
- Department of pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, PO BOX 3354, Riyadh, 11211, Kingdom of Saudi Arabia
| |
Collapse
|
52
|
Abstract
Epigenetic alterations such as DNA methylation defects and aberrant covalent histone modifications occur within all cancers and are selected for throughout the natural history of tumor formation, with changes being detectable in early onset, progression, and ultimately recurrence and metastasis. The ascertainment and use of these marks to identify at-risk patient populations, refine diagnostic criteria, and provide prognostic and predictive factors to guide treatment decisions are of growing clinical relevance. Furthermore, the targetable nature of epigenetic modifications provides a unique opportunity to alter treatment paradigms and provide new therapeutic options for patients whose malignancies possess these aberrant epigenetic modifications, paving the way for new and personalized medicine. DNA methylation has proven to be of significant clinical utility for its stability and relative ease of testing. The intent of this review is to elaborate upon well-supported examples of epigenetic precision medicine and how the field is moving forward, primarily in the context of aberrant DNA methylation.
Collapse
Affiliation(s)
- Rachael J Werner
- From the *Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | | | | |
Collapse
|
53
|
Abstract
PURPOSE OF REVIEW Prostate cancer is a disease of the elderly but a clinically relevant subset occurs early in life. In the current review, we discuss recent findings and the current understanding of the molecular underpinnings associated with early-onset prostate cancer (PCa) and the evidence supporting age-specific differences in the cancer genomes. RECENT FINDINGS Recent surveys of PCa patient cohorts have provided novel age-dependent links between germline and somatic aberrations which points to differences in the molecular cause and treatment options. SUMMARY Identifying the earliest molecular alterations in PCa can provide insight into the cause of the disease and biomarkers for patient risk stratification. Genomic aberrations of early-onset PCas display several patterns distinct from late-onset PCa genomes, suggesting age-dependent pathomechanisms involving alterations in the androgen receptor pathway.
Collapse
|
54
|
Böck J, Appenzeller S, Haertle L, Schneider T, Gehrig A, Schröder J, Rost S, Wolf B, Bartram CR, Sutter C, Haaf T. Single CpG hypermethylation, allele methylation errors, and decreased expression of multiple tumor suppressor genes in normal body cells of mutation-negative early-onset and high-risk breast cancer patients. Int J Cancer 2018; 143:1416-1425. [PMID: 29659014 PMCID: PMC6099327 DOI: 10.1002/ijc.31526] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 03/28/2018] [Indexed: 01/01/2023]
Abstract
To evaluate the role of constitutive epigenetic changes in normal body cells of BRCA1/BRCA2-mutation negative patients, we have developed a deep bisulfite sequencing assay targeting the promoter regions of 8 tumor suppressor (TS) genes (BRCA1, BRCA2, RAD51C, ATM, PTEN, TP53, MLH1, RB1) and the estrogene receptor gene (ESR1), which plays a role in tumor progression. We analyzed blood samples of two breast cancer (BC) cohorts with early onset (EO) and high risk (HR) for a heterozygous mutation, respectively, along with age-matched controls. Methylation analysis of up to 50,000 individual DNA molecules per gene and sample allowed quantification of epimutations (alleles with >50% methylated CpGs), which are associated with epigenetic silencing. Compared to ESR1, which is representative for an average promoter, TS genes were characterized by a very low (< 1%) average methylation level and a very low mean epimutation rate (EMR; < 0.0001% to 0.1%). With exception of BRCA1, which showed an increased EMR in BC (0.31% vs. 0.06%), there was no significant difference between patients and controls. One of 36 HR BC patients exhibited a dramatically increased EMR (14.7%) in BRCA1, consistent with a disease-causing epimutation. Approximately one third (15 of 44) EO BC patients exhibited increased rates of single CpG methylation errors in multiple TS genes. Both EO and HR BC patients exhibited global underexpression of blood TS genes. We propose that epigenetic abnormalities in normal body cells are indicative of disturbed mechanisms for maintaining low methylation and appropriate expression levels and may be associated with an increased BC risk.
Collapse
Affiliation(s)
- Julia Böck
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | | | - Larissa Haertle
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Tamara Schneider
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Andrea Gehrig
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Jörg Schröder
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Simone Rost
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Beat Wolf
- University of Applied Sciences Western Switzerland, Fribourg, Switzerland.,Department of Bioinformatics, University of Würzburg, Würzburg, Germany
| | - Claus R Bartram
- Institute of Human Genetics, University Hospital, Heidelberg, Germany
| | - Christian Sutter
- Institute of Human Genetics, University Hospital, Heidelberg, Germany
| | - Thomas Haaf
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| |
Collapse
|
55
|
Diversity of genetic events associated with MLH1 promoter methylation in Lynch syndrome families with heritable constitutional epimutation. Genet Med 2018; 20:1589-1599. [DOI: 10.1038/gim.2018.47] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 02/20/2018] [Indexed: 02/07/2023] Open
|
56
|
Saif I, Kasmi Y, Allali K, Ennaji MM. Prediction of DNA methylation in the promoter of gene suppressor tumor. Gene 2018; 651:166-173. [DOI: 10.1016/j.gene.2018.01.082] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 12/23/2017] [Accepted: 01/25/2018] [Indexed: 10/18/2022]
|
57
|
Joo JE, Dowty JG, Milne RL, Wong EM, Dugué PA, English D, Hopper JL, Goldgar DE, Giles GG, Southey MC. Heritable DNA methylation marks associated with susceptibility to breast cancer. Nat Commun 2018; 9:867. [PMID: 29491469 PMCID: PMC5830448 DOI: 10.1038/s41467-018-03058-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 01/08/2018] [Indexed: 02/03/2023] Open
Abstract
Mendelian-like inheritance of germline DNA methylation in cancer susceptibility genes has been previously reported. We aimed to scan the genome for heritable methylation marks associated with breast cancer susceptibility by studying 25 Australian multiple-case breast cancer families. Here we report genome-wide DNA methylation measured in 210 peripheral blood DNA samples provided by family members using the Infinium HumanMethylation450. We develop and apply a new statistical method to identify heritable methylation marks based on complex segregation analysis. We estimate carrier probabilities for the 1000 most heritable methylation marks based on family structure, and we use Cox proportional hazards survival analysis to identify 24 methylation marks with corresponding carrier probabilities significantly associated with breast cancer. We replicate an association with breast cancer risk for four of the 24 marks using an independent nested case-control study. Here, we report a novel approach for identifying heritable DNA methylation marks associated with breast cancer risk.
Collapse
Affiliation(s)
- Jihoon E Joo
- Department of Pathology, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, 3168, Australia
| | - James G Dowty
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Roger L Milne
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, VIC, 3004, Australia
| | - Ee Ming Wong
- Department of Pathology, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, 3168, Australia
| | - Pierre-Antoine Dugué
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, VIC, 3004, Australia
| | - Dallas English
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, VIC, 3004, Australia
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - David E Goldgar
- Department of Pathology, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Huntsman Cancer Institute, Salt Lake, UT, 84112, USA
| | - Graham G Giles
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, VIC, 3004, Australia
| | - Melissa C Southey
- Department of Pathology, The University of Melbourne, Melbourne, VIC, 3010, Australia.
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, 3168, Australia.
| |
Collapse
|
58
|
Liu X, Jiang Y, Nowak B, Qiang B, Cheng N, Chen Y, Plunkett W. Targeting BRCA1/2 deficient ovarian cancer with CNDAC-based drug combinations. Cancer Chemother Pharmacol 2018; 81:255-267. [PMID: 29189915 PMCID: PMC5777892 DOI: 10.1007/s00280-017-3483-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 11/15/2017] [Indexed: 12/18/2022]
Abstract
PURPOSE The mechanism of action of CNDAC (2'-C-cyano-2'-deoxy-1-β-D-arabino-pentofuranosyl-cytosine) is unique among deoxycytidine analogs because upon incorporation into DNA it causes a single strand break which is converted to a double strand break after DNA replication. This lesion requires homologous recombination (HR) for repair. CNDAC, as the parent nucleoside, DFP10917, and as an oral prodrug, sapacitabine, are undergoing clinical trials for hematological malignancies and solid tumors. The purpose of this study is to investigate the potential of CNDAC for the therapy of ovarian cancer (OC). METHODS Drug sensitivity was evaluated using a clonogenic survival assay. Drug combination effects were quantified by median effect analysis. RESULTS OC cells lacking function of the key HR genes, BRCA1 or BRCA2, were more sensitive to CNDAC than corresponding HR proficient cells. The sensitization was associated with greater levels of DNA damage in response to CNDAC at clinically achievable concentrations, manifested as chromosomal aberrations. Three classes of CNDAC-based drug combinations were investigated. First, the PARP1 inhibitors, rucaparib and talazoparib, were selectively synergistic with CNDAC in BRCA1/2 deficient OC cells (combination index < 1) at a relatively low concentration range. Second, cisplatin and oxaliplatin had additive combination effects with CNDAC (combination index ~ 1). Finally, paclitaxel and docetaxel achieved additive cell-killing effects with CNDAC at concentration ranges of the taxanes similar for both BRCA1/2 deficient and proficient OC cells. CONCLUSIONS This study provides mechanistic rationales for combining CNDAC with PARP inhibitors, platinum compounds and taxanes in ovarian cancer lacking BRCA1/2 function.
Collapse
Affiliation(s)
- Xiaojun Liu
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, 1901 East Road, 77054, Houston, TX, USA
| | - Yingjun Jiang
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, 1901 East Road, 77054, Houston, TX, USA
| | - Billie Nowak
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, 1901 East Road, 77054, Houston, TX, USA
| | - Bethany Qiang
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, 1901 East Road, 77054, Houston, TX, USA
| | - Nancy Cheng
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, 1901 East Road, 77054, Houston, TX, USA
| | - Yuling Chen
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, 1901 East Road, 77054, Houston, TX, USA
| | - William Plunkett
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, 1901 East Road, 77054, Houston, TX, USA.
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, 77030, Houston, TX, USA.
| |
Collapse
|
59
|
Dos Santos ES, Caputo SM, Castera L, Gendrot M, Briaux A, Breault M, Krieger S, Rogan PK, Mucaki EJ, Burke LJ, Bièche I, Houdayer C, Vaur D, Stoppa-Lyonnet D, Brown MA, Lallemand F, Rouleau E. Assessment of the functional impact of germline BRCA1/2 variants located in non-coding regions in families with breast and/or ovarian cancer predisposition. Breast Cancer Res Treat 2017; 168:311-325. [PMID: 29236234 DOI: 10.1007/s10549-017-4602-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 11/28/2017] [Indexed: 12/19/2022]
Abstract
PURPOSE The molecular mechanism of breast and/or ovarian cancer susceptibility remains unclear in the majority of patients. While germline mutations in the regulatory non-coding regions of BRCA1 and BRCA2 genes have been described, screening has generally been limited to coding regions. The aim of this study was to evaluate the contribution of BRCA1/2 non-coding variants. METHODS Four BRCA1/2 non-coding regions were screened using high-resolution melting analysis/Sanger sequencing or next-generation sequencing on DNA extracted from index cases with breast and ovarian cancer predisposition (3926 for BRCA1 and 3910 for BRCA2). The impact of a set of variants on BRCA1/2 gene regulation was evaluated by site-directed mutagenesis, transfection, followed by Luciferase gene reporter assay. RESULTS We identified a total of 117 variants and tested twelve BRCA1 and 8 BRCA2 variants mapping to promoter and intronic regions. We highlighted two neighboring BRCA1 promoter variants (c.-130del; c.-125C > T) and one BRCA2 promoter variants (c.-296C > T) inhibiting significantly the promoter activity. In the functional assays, a regulating region within the intron 12 was found with the same enhancing impact as within the intron 2. Furthermore, the variants c.81-3980A > G and c.4186-2022C > T suppress the positive effect of the introns 2 and 12, respectively, on the BRCA1 promoter activity. We also found some variants inducing the promoter activities. CONCLUSION In this study, we highlighted some variants among many, modulating negatively the promoter activity of BRCA1 or 2 and thus having a potential impact on the risk of developing cancer. This selection makes it possible to conduct future validation studies on a limited number of variants.
Collapse
Affiliation(s)
- E Santana Dos Santos
- Department of Oncology, Center for Translational Oncology, Cancer Institute of the State of São Paulo - ICESP, São Paulo, Brazil
- Service de Génétique, Institut Curie, Paris, France
- A.C.Camargo Cancer Center, São Paulo, Brazil
| | - S M Caputo
- Service de Génétique, Institut Curie, Paris, France
| | - L Castera
- Laboratoire de Biologie et de Génétique du Cancer, CLCC François Baclesse, INSERM 1079 Centre Normand de Génomique et de MédecinePersonnalisée, Caen, France
| | - M Gendrot
- Service de Génétique, Institut Curie, Paris, France
| | - A Briaux
- Service de Génétique, Institut Curie, Paris, France
| | - M Breault
- Service de Génétique, Institut Curie, Paris, France
| | - S Krieger
- Laboratoire de Biologie et de Génétique du Cancer, CLCC François Baclesse, INSERM 1079 Centre Normand de Génomique et de MédecinePersonnalisée, Caen, France
| | - P K Rogan
- Department of Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - E J Mucaki
- Department of Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - L J Burke
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - I Bièche
- Service de Génétique, Institut Curie, Paris, France
- Université Paris Descartes, Paris, France
| | - C Houdayer
- Service de Génétique, Institut Curie, Paris, France
- Université Paris Descartes, Paris, France
| | - D Vaur
- Laboratoire de Biologie et de Génétique du Cancer, CLCC François Baclesse, INSERM 1079 Centre Normand de Génomique et de MédecinePersonnalisée, Caen, France
| | - D Stoppa-Lyonnet
- Service de Génétique, Institut Curie, Paris, France
- Université Paris Descartes, Paris, France
| | - M A Brown
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - F Lallemand
- Service de Génétique, Institut Curie, Paris, France.
| | | |
Collapse
|
60
|
Christmann M, Kaina B. Epigenetic regulation of DNA repair genes and implications for tumor therapy. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2017; 780:15-28. [PMID: 31395346 DOI: 10.1016/j.mrrev.2017.10.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 10/05/2017] [Accepted: 10/06/2017] [Indexed: 12/31/2022]
Abstract
DNA repair represents the first barrier against genotoxic stress causing metabolic changes, inflammation and cancer. Besides its role in preventing cancer, DNA repair needs also to be considered during cancer treatment with radiation and DNA damaging drugs as it impacts therapy outcome. The DNA repair capacity is mainly governed by the expression level of repair genes. Alterations in the expression of repair genes can occur due to mutations in their coding or promoter region, changes in the expression of transcription factors activating or repressing these genes, and/or epigenetic factors changing histone modifications and CpG promoter methylation or demethylation levels. In this review we provide an overview on the epigenetic regulation of DNA repair genes. We summarize the mechanisms underlying CpG methylation and demethylation, with de novo methyltransferases and DNA repair involved in gain and loss of CpG methylation, respectively. We discuss the role of components of the DNA damage response, p53, PARP-1 and GADD45a on the regulation of the DNA (cytosine-5)-methyltransferase DNMT1, the key enzyme responsible for gene silencing. We stress the relevance of epigenetic silencing of DNA repair genes for tumor formation and tumor therapy. A paradigmatic example is provided by the DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT), which is silenced in up to 40% of various cancers through CpG promoter methylation. The CpG methylation status of the MGMT promoter strongly correlates with clinical outcome and, therefore, is used as prognostic marker during glioblastoma therapy. Mismatch repair genes are also subject of epigenetic silencing, which was shown to correlate with colorectal cancer formation. For many other repair genes shown to be epigenetically regulated the clinical outcome is not yet clear. We also address the question of whether genotoxic stress itself can lead to epigenetic alterations of genes encoding proteins involved in the defense against genotoxic stress.
Collapse
Affiliation(s)
- Markus Christmann
- Department of Toxicology, University of Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany.
| | - Bernd Kaina
- Department of Toxicology, University of Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany.
| |
Collapse
|
61
|
Lieberman R, You M. Corrupting the DNA damage response: a critical role for Rad52 in tumor cell survival. Aging (Albany NY) 2017; 9:1647-1659. [PMID: 28722656 PMCID: PMC5559167 DOI: 10.18632/aging.101263] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 07/12/2017] [Indexed: 12/31/2022]
Abstract
The DNA damage response enables cells to survive, maintain genome integrity, and to safeguard the transmission of high-fidelity genetic information. Upon sensing DNA damage, cells respond by activating this multi-faceted DNA damage response leading to restoration of the cell, senescence, programmed cell death, or genomic instability if the cell survives without proper repair. However, unlike normal cells, cancer cells maintain a marked level of genomic instability. Because of this enhanced propensity to accumulate DNA damage, tumor cells rely on homologous recombination repair as a means of protection from the lethal effect of both spontaneous and therapy-induced double-strand breaks (DSBs) in DNA. Thus, modulation of DNA repair pathways have important consequences for genomic instability within tumor cell biology and viability maintenance under high genotoxic stress. Efforts are underway to manipulate specific components of the DNA damage response in order to selectively induce tumor cell death by augmenting genomic instability past a viable threshold. New evidence suggests that RAD52, a component of the homologous recombination pathway, is important for the maintenance of tumor genome integrity. This review highlights recent reports indicating that reducing homologous recombination through inhibition of RAD52 may represent an important focus for cancer therapy and the specific efforts that are already demonstrating potential.
Collapse
Affiliation(s)
- Rachel Lieberman
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ming You
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
62
|
Khakpour G, Noruzinia M, Izadi P, Karami F, Ahmadvand M, Heshmat R, Amoli MM, Tavakkoly-Bazzaz J. Methylomics of breast cancer: Seeking epimarkers in peripheral blood of young subjects. Tumour Biol 2017; 39:1010428317695040. [PMID: 28349825 DOI: 10.1177/1010428317695040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Critical roles of epigenomic alterations in the pathogenesis of breast cancer have recently seized great attentions toward finding epimarkers in either non-invasive or semi-non-invasive samples as well as peripheral blood. In this way, methylated DNA immunoprecipitation microarray (MeDIP-chip) was performed on DNA samples isolated from white blood cells of 30 breast cancer patients compared to 30 healthy controls. A total of 1799 differentially methylated regions were identified including SLC6A3, Rab40C, ZNF584, and FOXD3 whose significant methylation differences were confirmed in breast cancer patients through quantitative real-time polymerase chain reaction. Hypermethylation of APC, HDAC1, and GSK1 genes has been previously reported in more than one study on tissue samples of breast cancer. Methylation of those aforementioned genes in white blood cells of our young patients not only relies on their importance in breast cancer pathogenesis but also may highlight their potential as early epimarkers that makes further assessments necessary in large cohort studies.
Collapse
Affiliation(s)
- Golnaz Khakpour
- 1 Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Noruzinia
- 2 Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Pantea Izadi
- 1 Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Karami
- 3 Department of Medical Genetics, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Ahmadvand
- 4 Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramin Heshmat
- 5 Chronic Disease Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa M Amoli
- 6 Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Javad Tavakkoly-Bazzaz
- 1 Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
63
|
Ha JR, Siegel PM, Ursini-Siegel J. The Tyrosine Kinome Dictates Breast Cancer Heterogeneity and Therapeutic Responsiveness. J Cell Biochem 2016; 117:1971-90. [PMID: 27392311 DOI: 10.1002/jcb.25561] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 03/24/2016] [Indexed: 12/13/2022]
Abstract
Phospho-tyrosine signaling networks control numerous biological processes including cellular differentiation, cell growth and survival, motility, and invasion. Aberrant regulation of the tyrosine kinome is a hallmark of malignancy and influences all stages of breast cancer progression, from initiation to the development of metastatic disease. The success of specific tyrosine kinase inhibitors strongly validates the clinical relevance of tyrosine phosphorylation networks in breast cancer pathology. However, a significant degree of redundancy exists within the tyrosine kinome. Numerous receptor and cytoplasmic tyrosine kinases converge on a core set of signaling regulators, including adaptor proteins and tyrosine phosphatases, to amplify pro-tumorigenic signal transduction pathways. Mutational activation, amplification, or overexpression of one or more components of the tyrosine kinome represents key contributing events responsible for the tumor heterogeneity that is observed in breast cancers. It is this molecular heterogeneity that has become the most significant barrier to durable clinical responses due to the development of therapeutic resistance. This review focuses on recent literature that supports a prominent role for specific components of the tyrosine kinome in the emergence of unique breast cancer subtypes and in shaping breast cancer plasticity, sensitivity to targeted therapies, and the eventual emergence of acquired resistance. J. Cell. Biochem. 117: 1971-1990, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jacqueline R Ha
- Lady Davis Institute for Medical Research, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada.,Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - Peter M Siegel
- Department of Medicine, McGill University, Montreal, Quebec, Canada.,Department of Oncology, McGill University, Montreal, Quebec, Canada.,Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada.,Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | - Josie Ursini-Siegel
- Lady Davis Institute for Medical Research, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada.,Department of Oncology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
64
|
Etzold A, Galetzka D, Weis E, Bartsch O, Haaf T, Spix C, Itzel T, Schweiger S, Strand D, Strand S, Zechner U. CAF-like state in primary skin fibroblasts with constitutional BRCA1 epimutation sheds new light on tumor suppressor deficiency-related changes in healthy tissue. Epigenetics 2016; 11:120-31. [PMID: 26949839 DOI: 10.1080/15592294.2016.1140295] [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: 01/03/2023] Open
Abstract
Constitutive epimutations of tumor suppressor genes are increasingly considered as cancer predisposing factors equally to sequence mutations. In light of the emerging role of the microenvironment for cancer predisposition, initiation, and progression, we aimed to characterize the consequences of a BRCA1 epimutation in cells of mesenchymal origin. We performed a comprehensive molecular and cellular comparison of primary dermal fibroblasts taken from a monozygous twin pair discordant for recurrent cancers and BRCA1 epimutation, whose exceptional clinical case we previously reported in this journal. Comparative transcriptome analysis identified differential expression of extracellular matrix-related genes and pro-tumorigenic growth factors, such as collagens and CXC chemokines. Moreover, genes known to be key markers of so called cancer-associated fibroblasts (CAFs), such as ACTA2, FAP, PDPN, and TNC, were upregulated in fibroblasts of the affected twin (BRCA1(mosMe)) in comparison to those of the healthy twin (BRCA1(wt)). Further analyses detected CAF-typical cellular features, including an elevated growth rate, enhanced migration, altered actin architecture and increased production of ketone bodies in BRCA1(mosMe) fibroblasts compared to BRCA1(wt) fibroblasts. In addition, conditioned medium of BRCA1(mosMe) fibroblasts was more potent than conditioned medium of BRCA1(wt) fibroblasts to promote cell proliferation in an epithelial and a cancer cell line. Our data demonstrate, that a CAF-like state is not an exclusive feature of tumor-associated tissue but also exists in healthy tissue with tumor suppressor deficiency. The naturally occurring phenomenon of twin fibroblasts differing in their BRCA1 methylation status revealed to be a unique powerful tool for exploring tumor suppressor deficiency-related changes in healthy tissue, reinforcing their significance for cancer predisposition.
Collapse
Affiliation(s)
- Anna Etzold
- a Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz , Mainz , Germany
| | - Danuta Galetzka
- a Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz , Mainz , Germany
| | - Eva Weis
- a Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz , Mainz , Germany
| | - Oliver Bartsch
- a Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz , Mainz , Germany
| | - Thomas Haaf
- b Institute of Human Genetics, Julius Maximilians University , Würzburg , Germany
| | - Claudia Spix
- c Institute of Medical Biometry, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University Mainz , Mainz , Germany
| | - Timo Itzel
- c Institute of Medical Biometry, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University Mainz , Mainz , Germany
| | - Susann Schweiger
- a Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz , Mainz , Germany
| | - Dennis Strand
- d First Department of Internal Medicine , University Medical Center of the Johannes Gutenberg University Mainz , Mainz , Germany
| | - Susanne Strand
- d First Department of Internal Medicine , University Medical Center of the Johannes Gutenberg University Mainz , Mainz , Germany
| | - Ulrich Zechner
- a Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz , Mainz , Germany
| |
Collapse
|
65
|
Kalvala A, Gao L, Aguila B, Reese T, Otterson GA, Villalona-Calero MA, Duan W. Overexpression of Rad51C splice variants in colorectal tumors. Oncotarget 2016; 6:8777-87. [PMID: 25669972 PMCID: PMC4496183 DOI: 10.18632/oncotarget.3209] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 12/24/2014] [Indexed: 01/04/2023] Open
Abstract
Functional alterations in Rad51C are the cause of the Fanconi anemia complementation group O (FANCO) gene disorder. We have identified novel splice variants of Rad51C mRNA in colorectal tumors and cells. The alternatively spliced transcript variants are formed either without exon-7 (variant 1), without exon 6 and 7 (variant 2) or without exon 7 and 8 (variant 3). Real time PCR analysis of nine pair-matched colorectal tumors and non-tumors showed that variant 1 was overexpressed in tumors compared to matched non-tumors. Among 38 colorectal tumor RNA samples analyzed, 18 contained variant 1, 12 contained variant 2, 14 contained variant 3, and eight expressed full length Rad51C exclusively. Bisulfite DNA sequencing showed promoter methylation of Rad51C in tumor cells. 5-azacytidine treatment of LS-174T cells caused a 14 fold increase in variant 1, a 4.8 fold increase for variant 3 and 3.4 fold for variant 2 compared to 2.5 fold increase in WT. Expression of Rad51C variants is associated with FANCD2 foci positive colorectal tumors and is associated with microsatellite stability in those tumors. Further investigation is needed to elucidate differential function of the Rad51C variants to evaluate potential effects in drug resistance and DNA repair.
Collapse
Affiliation(s)
- Arjun Kalvala
- Comprehensive Cancer Center, The Ohio State University College of Medicine and Public Health, Columbus, Ohio, U.S.A
| | - Li Gao
- Comprehensive Cancer Center, The Ohio State University College of Medicine and Public Health, Columbus, Ohio, U.S.A
| | - Brittany Aguila
- Comprehensive Cancer Center, The Ohio State University College of Medicine and Public Health, Columbus, Ohio, U.S.A
| | - Tyler Reese
- Comprehensive Cancer Center, The Ohio State University College of Medicine and Public Health, Columbus, Ohio, U.S.A
| | - Gregory A Otterson
- Comprehensive Cancer Center, The Ohio State University College of Medicine and Public Health, Columbus, Ohio, U.S.A.,Division of Medical Oncology Department of Internal Medicine, The Ohio State University College of Medicine and Public Health, Columbus, Ohio, U.S.A
| | - Miguel A Villalona-Calero
- Comprehensive Cancer Center, The Ohio State University College of Medicine and Public Health, Columbus, Ohio, U.S.A.,Division of Medical Oncology Department of Internal Medicine, The Ohio State University College of Medicine and Public Health, Columbus, Ohio, U.S.A.,Department of Pharmacology at The Ohio State University College of Medicine and Public Health, Columbus, Ohio, U.S.A
| | - Wenrui Duan
- Comprehensive Cancer Center, The Ohio State University College of Medicine and Public Health, Columbus, Ohio, U.S.A.,Division of Medical Oncology Department of Internal Medicine, The Ohio State University College of Medicine and Public Health, Columbus, Ohio, U.S.A
| |
Collapse
|
66
|
Huang F, Goyal N, Sullivan K, Hanamshet K, Patel M, Mazina OM, Wang CX, An WF, Spoonamore J, Metkar S, Emmitte KA, Cocklin S, Skorski T, Mazin AV. Targeting BRCA1- and BRCA2-deficient cells with RAD52 small molecule inhibitors. Nucleic Acids Res 2016; 44:4189-99. [PMID: 26873923 PMCID: PMC4872086 DOI: 10.1093/nar/gkw087] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 02/03/2016] [Indexed: 12/20/2022] Open
Abstract
RAD52 is a member of the homologous recombination (HR) pathway that is important for maintenance of genome integrity. While single RAD52 mutations show no significant phenotype in mammals, their combination with mutations in genes that cause hereditary breast cancer and ovarian cancer like BRCA1, BRCA2, PALB2 and RAD51C are lethal. Consequently, RAD52 may represent an important target for cancer therapy. In vitro, RAD52 has ssDNA annealing and DNA strand exchange activities. Here, to identify small molecule inhibitors of RAD52 we screened a 372,903-compound library using a fluorescence-quenching assay for ssDNA annealing activity of RAD52. The obtained 70 putative inhibitors were further characterized using biochemical and cell-based assays. As a result, we identified compounds that specifically inhibit the biochemical activities of RAD52, suppress growth of BRCA1- and BRCA2-deficient cells and inhibit RAD52-dependent single-strand annealing (SSA) in human cells. We will use these compounds for development of novel cancer therapy and as a probe to study mechanisms of DNA repair.
Collapse
Affiliation(s)
- Fei Huang
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Nadish Goyal
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Katherine Sullivan
- Department of Microbiology and Immunology, and Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 10140, USA
| | - Kritika Hanamshet
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Mikir Patel
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Olga M Mazina
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Charles X Wang
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - W Frank An
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - James Spoonamore
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Shailesh Metkar
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Kyle A Emmitte
- Vanderbilt Specialized Chemistry Center for Accelerated Probe Development, Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology and Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Simon Cocklin
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Tomasz Skorski
- Department of Microbiology and Immunology, and Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 10140, USA
| | - Alexander V Mazin
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| |
Collapse
|
67
|
Abstract
Genetic causes for human disorders are being discovered at an unprecedented pace. A growing subclass of disease-causing mutations involves changes in the epigenome or in the abundance and activity of proteins that regulate chromatin structure. This article focuses on research that has uncovered human diseases that stem from such epigenetic deregulation. Disease may be caused by direct changes in epigenetic marks, such as DNA methylation, commonly found to affect imprinted gene regulation. Also described are disease-causing genetic mutations in epigenetic modifiers that either affect chromatin in trans or have a cis effect in altering chromatin configuration.
Collapse
Affiliation(s)
- Huda Y Zoghbi
- Howard Hughes Medical Institute, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Arthur L Beaudet
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
68
|
Affiliation(s)
- Krisztian Homicsko
- Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - George Coukos
- Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; and Ludwig Institute Branch at the University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
69
|
Abstract
Constitutional epimutation, which is an aberration in gene expression due to an altered epigenotype that is widely distributed in normal tissues (albeit frequently mosaic), provides an alternative mechanism to genetic mutation for cancer predisposition. Observational studies in cancer-affected families have revealed intergenerational inheritance of constitutional epimutation, providing unique insights into the heritability of epigenetic traits in humans. In this Opinion article, the potential contribution of constitutional epimutation to the 'missing' causality and heritability of cancer is explored.
Collapse
Affiliation(s)
- Megan P Hitchins
- Department of Medicine (Oncology), Stanford Cancer Institute, Stanford University, Grant Building S169, 1291 Welch Road, Stanford, California 94305, USA
| |
Collapse
|
70
|
Manié E, Popova T, Battistella A, Tarabeux J, Caux-Moncoutier V, Golmard L, Smith NK, Mueller CR, Mariani O, Sigal-Zafrani B, Dubois T, Vincent-Salomon A, Houdayer C, Stoppa-Lyonnet D, Stern MH. Genomic hallmarks of homologous recombination deficiency in invasive breast carcinomas. Int J Cancer 2015; 138:891-900. [DOI: 10.1002/ijc.29829] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 06/25/2015] [Accepted: 07/30/2015] [Indexed: 01/09/2023]
Affiliation(s)
- Elodie Manié
- Centre De Recherche; Institut Curie; Paris F-75248 France
- INSERM U830; Paris F-75248 France
| | - Tatiana Popova
- Centre De Recherche; Institut Curie; Paris F-75248 France
- INSERM U830; Paris F-75248 France
| | - Aude Battistella
- Centre De Recherche; Institut Curie; Paris F-75248 France
- INSERM U830; Paris F-75248 France
| | - Julien Tarabeux
- Centre De Recherche; Institut Curie; Paris F-75248 France
- INSERM U830; Paris F-75248 France
| | | | - Lisa Golmard
- Centre De Recherche; Institut Curie; Paris F-75248 France
- INSERM U830; Paris F-75248 France
- Département De Biologie Des Tumeurs; Institut Curie; Paris F-75248 France
| | - Nicholas K. Smith
- Centre De Recherche; Institut Curie; Paris F-75248 France
- INSERM U830; Paris F-75248 France
| | - Christopher R. Mueller
- Centre De Recherche; Institut Curie; Paris F-75248 France
- INSERM U830; Paris F-75248 France
- Queen's Cancer Research Institute, Queen's University, Kingston; Ontario K7L 3N6 Canada
| | - Odette Mariani
- Département De Biologie Des Tumeurs; Institut Curie; Paris F-75248 France
- Centre De Ressources Biologiques; Institut Curie; Paris F-75248 France
| | | | - Thierry Dubois
- Centre De Recherche; Institut Curie; Paris F-75248 France
- Département De Recherche Translationnelle; Institut Curie; Paris F-75248 France
| | | | - Claude Houdayer
- Centre De Recherche; Institut Curie; Paris F-75248 France
- INSERM U830; Paris F-75248 France
- Département De Biologie Des Tumeurs; Institut Curie; Paris F-75248 France
| | - Dominique Stoppa-Lyonnet
- Centre De Recherche; Institut Curie; Paris F-75248 France
- INSERM U830; Paris F-75248 France
- Département De Biologie Des Tumeurs; Institut Curie; Paris F-75248 France
- Sorbonne Paris Cité; University Paris-Descartes; Paris F-75270 France
| | - Marc-Henri Stern
- Centre De Recherche; Institut Curie; Paris F-75248 France
- INSERM U830; Paris F-75248 France
| |
Collapse
|
71
|
Ding Y, Zou W, Zhu C, Min H, Ma D, Chen B, Ye M, Pan Y, Cao L, Wan Y, Zhu Q, Xia H, Zhang W, Feng Y, Gao Q, Yi L. Promoter methylation is not associated with FLCN irregulation in lung cyst lesions of primary spontaneous pneumothorax. Mol Med Rep 2015; 12:7770-6. [PMID: 26398834 DOI: 10.3892/mmr.2015.4341] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 08/25/2015] [Indexed: 11/05/2022] Open
Abstract
Germline mutations in FLCN are responsible for ~10% of patients with primary spontaneous pneumothorax (PSP), characterized by multiple lung cysts in the middle/lower lobes and recurrent pneumothorax. These clinical features are also observed in a substantial portion of patients with sporadic PSP exhibiting no FLCN coding mutations. To assess the potential underlying mechanisms, 71 patients with PSP were selected, including 69 sporadic and 2 familial cases, who bared FLCN mutation‑like lung cysts, however, harbored no FLCN protein‑altering mutations. Notably, in a significant proportion of the patients, FLCN irregulation was observed at the transcript and protein levels. Genetic analyses of the cis‑regulatory region of FLCN were performed by sequencing and multiplex ligation‑dependent probe amplification assay. No inheritable DNA defect was detected, with the exception of a heterozygous deletion spanning the FLCN promoter, which was identified in a family with PSP. This mutation caused a reduction in the expression of FLCN in the lung cysts. Pedigree analysis demonstrated that haploinsufficiency of FLCN was pathogenic. To determine whether epigenetic mechanisms may be involved in the irregulation of FLCN, the promoter methylation status was measured in the remainder of the patients. No evidence of FLCN promoter methylation was demonstrated. The present study suggested that FLCN irregulation in lung cysts of PSP is not associated with promoter methylation.
Collapse
Affiliation(s)
- Yibing Ding
- Center for Translational Medicine, Nanjing University Medical School, Nanjing, Jiangsu 210093, P.R. China
| | - Wei Zou
- Department of Thoracic Surgery, Nanjing Chest Hospital, Nanjing, Jiangsu 210093, P.R. China
| | - Chengchu Zhu
- Department of Cardiothoracic Surgery, Taizhou Hospital of Zhejiang, Wenzhou Medical University, Linhai, Zhejiang 317000, P.R. China
| | - Haiyan Min
- Center for Translational Medicine, Nanjing University Medical School, Nanjing, Jiangsu 210093, P.R. China
| | - Dehua Ma
- Department of Cardiothoracic Surgery, Taizhou Hospital of Zhejiang, Wenzhou Medical University, Linhai, Zhejiang 317000, P.R. China
| | - Baofu Chen
- Department of Cardiothoracic Surgery, Taizhou Hospital of Zhejiang, Wenzhou Medical University, Linhai, Zhejiang 317000, P.R. China
| | - Minhua Ye
- Department of Cardiothoracic Surgery, Taizhou Hospital of Zhejiang, Wenzhou Medical University, Linhai, Zhejiang 317000, P.R. China
| | - Yanqing Pan
- Department of Thoracic Surgery, Nanjing Chest Hospital, Nanjing, Jiangsu 210093, P.R. China
| | - Lei Cao
- Department of Thoracic Surgery, Nanjing Chest Hospital, Nanjing, Jiangsu 210093, P.R. China
| | - Yueming Wan
- Department of Thoracic Surgery, Nanjing Chest Hospital, Nanjing, Jiangsu 210093, P.R. China
| | - Qiuxiang Zhu
- Center for Translational Medicine, Nanjing University Medical School, Nanjing, Jiangsu 210093, P.R. China
| | - Haizhen Xia
- Center for Translational Medicine, Nanjing University Medical School, Nanjing, Jiangsu 210093, P.R. China
| | - Wenwen Zhang
- Center for Translational Medicine, Nanjing University Medical School, Nanjing, Jiangsu 210093, P.R. China
| | - Ying Feng
- Center for Translational Medicine, Nanjing University Medical School, Nanjing, Jiangsu 210093, P.R. China
| | - Qian Gao
- Center for Translational Medicine, Nanjing University Medical School, Nanjing, Jiangsu 210093, P.R. China
| | - Long Yi
- Center for Translational Medicine, Nanjing University Medical School, Nanjing, Jiangsu 210093, P.R. China
| |
Collapse
|
72
|
Pirouzpanah S, Taleban FA, Mehdipour P, Atri M. Association of folate and other one-carbon related nutrients with hypermethylation status and expression of RARB, BRCA1, and RASSF1A genes in breast cancer patients. J Mol Med (Berl) 2015; 93:917-934. [PMID: 25805039 DOI: 10.1007/s00109-015-1268-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Revised: 02/11/2015] [Accepted: 02/16/2015] [Indexed: 02/08/2023]
Abstract
UNLABELLED Dietary methyl group donors could influence the hypermethylation status of certain putative genes. The present study explored the possible associations of dietary intake of one-carbon metabolism-related nutrients with promoter hypermethylation status and expression of retinoic acid receptor-beta (RARB), breast cancer-1 (BRCA1), and Ras association domain family-1, isoform A (RASSF1A) genes in Iranian women with breast cancer (BC). The hypermethylation status was investigated in 146 dissected BC tissue samples using methylation-specific PCR. The expression level was evaluated by real-time RT-PCR. Dietary nutrients were estimated using a validated 136-item food frequency questionnaire. Expression levels of the genes were associated with the unmethylated status of related promoters (p < 0.05). The crude dietary folate and adjusted cobalamin intakes were inversely associated with methylated RARB and BRCA1. Low intake of residual folate and cobalamin was correlated with the methylated status of RARB for subjects at <48 years of age, and folate alone was linked to BRCA1 at >48 years of age. High dietary intake of riboflavin and pyridoxine was the only determinant of the methylated promoter of RARB at odds ratios (ORs) of 4.15 (95 % confidence interval (CI) 1.28-13.50) and 2.53 (95 % CI 1.14-3.83) in multivariate models, respectively. One-carbon nutrients most often correlated inversely with the methylation-influenced expression of RARB. Although high folate intake increased the chance of unmethylation-dependent overexpression of BRCA1 3-fold, cobalamin and methionine were inversely linked to methylation-mediated expression. Nutritional epigenomics less actively influenced RASSF1A. These findings provide new insights into and a basic understanding of the selective contributions of individual B vitamins on hypermethylation and methylation-related expression of RARB and BRCA1 in BC. KEY MESSAGE Hypermethylation at promoters of RARB, BRCA1, and RASSF1A is associated with reduced transcript levels of the respective gene in primary breast cancer tissue samples. Dietary folate and cobalamin intake is inversely associated with methylated RARB and BRCA1. High dietary intake of riboflavin and pyridoxine is associated with increased methylation in the RARB promoter. There is evidence for the age-dependent effects of nutrient intake on promoter methylation status. Bioavailability to the pool of nutrients might determine selectivity.
Collapse
Affiliation(s)
- Saeed Pirouzpanah
- Department of Biochemistry and Dietetics, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, 5166614711, Iran,
| | | | | | | |
Collapse
|
73
|
DNA methylation as a promising landscape: A simple blood test for breast cancer prediction. Tumour Biol 2015; 36:4905-12. [PMID: 26076810 DOI: 10.1007/s13277-015-3567-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 05/13/2015] [Indexed: 01/27/2023] Open
Abstract
Breast cancer is the most common malignancy among women worldwide. Risk assessment is one of the main services delivered by cancer clinics. Biomarker analysis on different tissues including the peripheral blood can provide crucial information. One of the potential epigenetic biomarkers (epimarkers) is introduced as the peripheral blood DNA methylation pattern. This study was conducted to evaluate the potential value of peripheral blood epimarkers as an accessible tool to predict the risk of breast cancer development. WBC's DNA was the focus of several case-control studies at both genome wide and candidate gene levels to reveal epigenetic changes accounting for predisposition to breast cancer, leading to suggest that ATM, TITF1, SFRP1, NUP155, NEUROD1, ZNF217, DBC2, DOK7 and ESR1 genes and the LINE1, Alu and Sat2 DNA elements could be considered as the potential epimarkers. To address that by which mechanisms WBC's DNA methylation patterns could be linked to the propensity to breast cancer, several contemplations have been offered. Constitutional epimutation during embryonic life, and methylation changes secondary to either environmental exposures or tumor-mediated immune response, are the two main mechanisms. One can deduce that epimarkers based on their potential properties or regulatory impacts on cancer-related genes may be employed for risk prediction, prognosis, and survival inferences that are highly required for breast cancer management toward personalized medicine.
Collapse
|
74
|
Pliushch G, Schneider E, Schneider T, El Hajj N, Rösner S, Strowitzki T, Haaf T. In vitro maturation of oocytes is not associated with altered deoxyribonucleic acid methylation patterns in children from in vitro fertilization or intracytoplasmic sperm injection. Fertil Steril 2015; 103:720-7.e1. [PMID: 25572872 DOI: 10.1016/j.fertnstert.2014.12.096] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 12/08/2014] [Accepted: 12/08/2014] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To study the possible transmission, to the next generation, of epigenetic defects associated with in vitro maturation (IVM) of human oocytes. DESIGN Case-control study using epigenetic data. SETTING Two collaborating university departments. PATIENT(S) Eleven IVM newborns and 19 controls, conceived by conventional assisted reproduction. INTERVENTION(S) Chorionic villus and cord-blood sampling. MAIN OUTCOME MEASURE(S) Using bisulfite pyrosequencing, we have measured average methylation levels of 6 imprinted (LIT1, MEG, MEST, NESPas, PEG3, and SNRPN), 5 tumor-suppressor (APC, ATM, BRCA1, RAD51C, and TP53), 2 pluripotency (NANOG and OCT4), and 2 metabolic (LEP and NR3C1) genes, as well as 2 repetitive elements (ALU and LINE1) in 2 tissues of IVM and control neonates. Using deep bisulfite sequencing, we have determined methylation patterns of many individual DNA molecules to detect rare RAD51C epimutations (allele methylation errors). RESULT(S) No statistically significant impact was found of IVM on chorionic villus and cord-blood DNA methylation at the studied developmentally important genes and interspersed repeats. The RAD51C epimutation rate was low (0.5% ± 0.1%) in all analyzed samples. CONCLUSION(S) IVM-induced epigenetic changes in offspring, if any, are relatively small in magnitude and/or infrequent.
Collapse
Affiliation(s)
- Galyna Pliushch
- Institute of Human Genetics, Julius Maximilians University, Wuerzburg, Germany
| | - Eberhard Schneider
- Institute of Human Genetics, Julius Maximilians University, Wuerzburg, Germany
| | - Tamara Schneider
- Institute of Human Genetics, Julius Maximilians University, Wuerzburg, Germany
| | - Nady El Hajj
- Institute of Human Genetics, Julius Maximilians University, Wuerzburg, Germany
| | - Sabine Rösner
- Department of Gynecological Endocrinology and Reproductive Medicine, Women's Hospital, Ruprecht Karls University, Heidelberg, Germany
| | - Thomas Strowitzki
- Department of Gynecological Endocrinology and Reproductive Medicine, Women's Hospital, Ruprecht Karls University, Heidelberg, Germany
| | - Thomas Haaf
- Institute of Human Genetics, Julius Maximilians University, Wuerzburg, Germany.
| |
Collapse
|
75
|
Al-Moghrabi N, Nofel A, Al-Yousef N, Madkhali S, Bin Amer SM, Alaiya A, Shinwari Z, Al-Tweigeri T, Karakas B, Tulbah A, Aboussekhra A. The molecular significance of methylated BRCA1 promoter in white blood cells of cancer-free females. BMC Cancer 2014; 14:830. [PMID: 25403427 PMCID: PMC4289167 DOI: 10.1186/1471-2407-14-830] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 10/23/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND BRCA1 promoter methylation has been detected in DNA from peripheral blood cells of both breast cancer patients and cancer-free females. However, the pathological significance of this epigenetic change in white blood cells (WBC) remains an open question. In this study, we hypothesized that if constitutional BRCA1 methylation reflects an elevated risk for developing breast cancer (BC), WBC that harbor methylated BRCA1 in both cancer-free females and BC patients should exhibit similar molecular changes. METHODS BRCA1 promoter methylation was examined by methylation-specific PCR in WBC from 155 breast cancer patients and 143 cancer-free females. The Human Breast Cancer EpiTect Methyl II Signature PCR Array and The Human Breast Cancer RT2 Profiler™ PCR Array were used to study the methylation status and the expression profile of several breast cancer-related genes, respectively. In addition, we used label-free MS-based technique to study protein expression in plasma. RESULTS We have shown that 14.2% of BC patients and 9.1% of cancer-free females (carriers) harbored methylated BRCA1 promoter in their WBC. Interestingly, 66.7% of patients harbored methylated BRCA1 promoter in both WBC and tumors. Importantly, we have shown the presence of epigenetic changes in 9 other BC-related genes in WBC of both patients and carriers. Additionally, BRCA1 and 15 other important cancer -related genes were found to be differentially expressed in WBC from patients and carriers as compared to controls. Furthermore, we have shown that the carriers exhibited a unique plasma protein pattern different from those of BC patients and controls, with 10 proteins similarly differentially expressed in patients and carriers as compared to controls. CONCLUSIONS The present results suggest the presence of a strong link between aberrant methylation of the BRCA1 promoter in WBC and breast cancer -related molecular changes, which indicate the potential predisposition of the carriers for developing breast cancer. This informs the potential use of the aberrant methylation of BRCA1 promoter in WBC as a powerful non-invasive molecular marker for detecting predisposed individuals at a very early age.
Collapse
Affiliation(s)
- Nisreen Al-Moghrabi
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, PO BOX 3354, 11211 Riyadh, Kingdom of Saudi Arabia
| | - Asmaa Nofel
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, PO BOX 3354, 11211 Riyadh, Kingdom of Saudi Arabia
| | - Nujoud Al-Yousef
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, PO BOX 3354, 11211 Riyadh, Kingdom of Saudi Arabia
| | - Safia Madkhali
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, PO BOX 3354, 11211 Riyadh, Kingdom of Saudi Arabia
| | - Suad M Bin Amer
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, PO BOX 3354, 11211 Riyadh, Kingdom of Saudi Arabia
| | - Ayodele Alaiya
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, PO BOX 3354, 11211 Riyadh, Kingdom of Saudi Arabia
| | - Zakia Shinwari
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, PO BOX 3354, 11211 Riyadh, Kingdom of Saudi Arabia
| | - Taher Al-Tweigeri
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, PO BOX 3354, 11211 Riyadh, Kingdom of Saudi Arabia
| | - Bedri Karakas
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, PO BOX 3354, 11211 Riyadh, Kingdom of Saudi Arabia
| | - Asma Tulbah
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, PO BOX 3354, 11211 Riyadh, Kingdom of Saudi Arabia
| | - Abdelilah Aboussekhra
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, PO BOX 3354, 11211 Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
76
|
Methylation of the BRCA1 promoter in peripheral blood DNA is associated with triple-negative and medullary breast cancer. Breast Cancer Res Treat 2014; 148:615-22. [PMID: 25376744 DOI: 10.1007/s10549-014-3179-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 10/23/2014] [Indexed: 12/19/2022]
Abstract
It has been proposed that methylation signatures in blood-derived DNA may correlate with cancer risk. In this study, we evaluated whether methylation of the promoter region of the BRCA1 gene detectable in DNA from peripheral blood cells is a risk factor for breast cancer, in particular for tumors with pathologic features characteristic for cancers with BRCA1 gene mutations. We conducted a case-control study of 66 breast cancer cases and 36 unaffected controls. Cases were triple-negative or of medullary histology, or both; 30 carried a constitutional BRCA1 mutation and 36 did not carry a mutation. Blood for DNA methylation analysis was taken within three months of diagnosis. Methylation of the promoter of the BRCA1 gene was measured in cases and controls using methylation-sensitive high-resolution melting (MS-HRM). A sample with any detectable level of methylation was considered to be positive. Methylation of the BRCA1 promoter was detected in 15 of 66 cases and in 2 of 36 controls (OR 5.0, p = 0.03). Methylation was present in 15 of 36 women with breast cancer and without germline BRCA1 mutation, but in none of 30 women with breast cancer and a germline mutation (p < 0.01). The association between methylation and breast cancer was restricted to women with no constitutional BRCA1 mutation (OR 12.1, p = 0.0006). Methylation of the promoter of the BRCA1 gene detectable in peripheral blood DNA may be a marker of increased susceptibility to triple-negative or medullary breast cancer.
Collapse
|
77
|
Haytural H, Yalcinkaya N, Akan G, Arikan S, Ozkok E, Cakmakoglu B, Yaylim I, Aydin M, Atalar F. Identification of a novel BRCA2 and CHEK2 A-C-G-C haplotype in Turkish patients affected with breast cancer. Asian Pac J Cancer Prev 2014; 14:3229-35. [PMID: 23803109 DOI: 10.7314/apjcp.2013.14.5.3229] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Many breast cancers are caused by certain rare and familial mutations in the high or moderate penetrance genes BRCA1, BRCA2 and CHEK2. The aim of this study was to examine the allele and genotype frequencies of seven mutations in BRCA1, BRCA2 and CHEK2 genes in breast cancer patients and to investigate their isolated and combined associations with breast cancer risk. METHODS We genotyped seven mutations in BRCA1, BRCA2 and CHEK2 genes and then analyzed single variations and haplotype associations in 106 breast cancer patients and 80 healthy controls. RESULTS We found significant associations in the analyses of CHEK2- 1100delC (p=0.001) and BRCA1-5382insC (p=0.021) mutations in breast cancer patients compared to controls. The highest risk was observed among breast cancer patients carrying both CHEK2-1100delC and BRCA2- Met784Val mutations (OR=0.093; 95%CI 0.021-0.423; p=0.001). We identified one previously undescribed BRCA2 and a CHEK2 four-marker haplotype of A-C-G-C which was overrepresented (?2=7.655; p=0.0057) in the patient group compared to controls. CONCLUSION In this study, we identified a previously undescribed BRCA2 and CHEK2 A-C-G-C haplotype in association with the breast cancer in our population. Our results further suggest that the CHEK2-1100delC mutation in combination with BRCA2-Met784Val may lead to an unexpected high risk which needs to be confirmed in larger cohorts in order to better understand their role in the development and prognosis of breast cancer.
Collapse
Affiliation(s)
- Hazal Haytural
- Department of Neuroscience, Institute for Experimental Medicine Research, Istanbul University, Istanbul, Turkey
| | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Pergoli L, Favero C, Ruth M. P, Tarantini L, Calista D, Cavalleri T, Angelici L, Consonni D, Pier A. B, Angela C. P, Maria T. L, Bollati V. Blood DNA methylation, nevi number, and the risk of melanoma. Melanoma Res 2014; 24:480-7. [PMID: 25026000 PMCID: PMC6857929 DOI: 10.1097/cmr.0000000000000112] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Germline mutations determining increased cutaneous malignant melanoma (CMM) risk have been identified in familial and sporadic CMM cases, but they account only for a small proportion of CMM cases. Recent evidence suggests that germline epimutations (e.g. DNA methylation alterations), which can be inherited similarly to genomic mutations and can be detected in normal body cells (including blood), might increase susceptibility to cancer. The aim of the study was to identify germline epimutations of genes that were found to be mutated in familial CMM (p16, p14, CDK4, MC1R, hTERT), immune and inflammatory genes (ICAM-1, TNFα), DNA mismatch repair gene (MLH1), and repetitive elements (ALU, LINE-1, HERV-w). We measured DNA methylation using bisulfite pyrosequencing in peripheral blood mononuclear cells from 167 CMM cases and 164 sex-matched and age-matched controls. We used multivariable logistic regression models to evaluate the association between methylation levels and CMM status or presence of dysplastic nevi. We found an association between the risk of CMM and peripheral blood mononuclear cell methylation levels of TNFα [odds ratio (OR)=1.11, 95% confidence interval (CI)=1.03-1.18], CDK4 (OR=0.76, 95% CI=0.64-0.91), and MLH1 (OR=1.12, 95% CI=1.02-1.22). In control participants, the risk of developing dysplastic nevi was associated with methylation levels of TNFα (OR=0.81, 95% CI=0.69-0.95), hTERT (OR=0.90, 95% CI=0.82-0.99), and ALU (OR=1.56, 95% CI=1.02-2.39). Epimutations in CMM susceptibility genes and in genes involved in response to oxidative damage are associated with the risk of developing CMM or dysplastic nevi. Further studies measuring methylation levels of these genes in prospectively collected samples are warranted to further elucidate their role in the development and progression of CMM.
Collapse
Affiliation(s)
- Laura Pergoli
- Center of Molecular and Genetic Epidemiology, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Chiara Favero
- Center of Molecular and Genetic Epidemiology, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Pfeiffer Ruth M.
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Letizia Tarantini
- Center of Molecular and Genetic Epidemiology, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | | | - Tommaso Cavalleri
- Center of Molecular and Genetic Epidemiology, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Laura Angelici
- Center of Molecular and Genetic Epidemiology, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Dario Consonni
- Epidemiology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Bertazzi Pier A.
- Center of Molecular and Genetic Epidemiology, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Epidemiology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Pesatori Angela C.
- Center of Molecular and Genetic Epidemiology, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Epidemiology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Landi Maria T.
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Valentina Bollati
- Center of Molecular and Genetic Epidemiology, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Epidemiology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
79
|
Van Neste L, Van Criekinge W. We are all individuals... bioinformatics in the personalized medicine era. Cell Oncol (Dordr) 2014; 38:29-37. [PMID: 25204962 DOI: 10.1007/s13402-014-0195-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2014] [Indexed: 12/16/2022] Open
Abstract
The medical landscape is evolving at a rapid pace, creating the opportunity for more personalized patient treatment and shifting the way healthcare is approached and thought about. With the availability of (epi)genome-wide, transcriptomic and proteogenomic profiling techniques detailed characterization of a disease at the level of the individual is now possible, offering the opportunity for truly tailored approaches for treatment and patient care. While improvements are still expected, the techniques and the basic analytical tools have reached a state that these can be efficiently deployed in both routine research and clinical practice. Still, some major challenges remain. Notably, holistic approaches, integrating data from several sources, e.g. genomic and epigenomic, will increase the understanding of the underlying biological concepts and provide insight into the causes, effects and effective solutions. However, creating and validating such a knowledge base, potentially for different levels of expertise, and integrating several data points into meaningful information is not trivial.
Collapse
Affiliation(s)
- Leander Van Neste
- Department of Pathology, School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands,
| | | |
Collapse
|
80
|
Ruscito I, Dimitrova D, Vasconcelos I, Gellhaus K, Schwachula T, Bellati F, Zeillinger R, Benedetti-Panici P, Vergote I, Mahner S, Cacsire-Tong D, Concin N, Darb-Esfahani S, Lambrechts S, Sehouli J, Olek S, Braicu EI. BRCA1 gene promoter methylation status in high-grade serous ovarian cancer patients--a study of the tumour Bank ovarian cancer (TOC) and ovarian cancer diagnosis consortium (OVCAD). Eur J Cancer 2014; 50:2090-8. [PMID: 24889916 DOI: 10.1016/j.ejca.2014.05.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/02/2014] [Accepted: 05/05/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Mutations in BRCA1/2 genes are involved in the pathogenesis of breast and ovarian cancer. Inactivation of these genes can also be mediated by hypermethylation of CpGs in the promoter regions. Aim of this study was to analyse the clinical impact of BRCA1 promoter gene methylation status in a homogenous cohort of high-grade serous ovarian cancer (HGSOC) patients. METHODS The cohort included 257 primary HGSOC patients treated by cytoreduction and platinum-based chemotherapy. DNA was extracted from fresh frozen tissue samples. BRCA1 gene promoter methylation rate was assessed using polymerase chain reaction (PCR). RESULTS 14.8% of patients presented hypermethylation within a selected region of the BRCA1 promoter. The rate of hypermethylation was significantly higher in younger patients (20.8% hypermethylation in the age group ⩽ 58 years versus 8.7% hypermethylation in the age group >58 years; p = 0.008). Optimal tumour debulking could be reached in 63% of patients, without significant differences in the extent of residual disease with respect to the methylation status. No impact of BRCA1 gene promoter methylation status on progression free- and overall-survival rates was found. No significant differences within BRCA1 promoter methylation status between primary and metastatic tissue could be observed. These results on BRCA1 promoter methylation status were also confirmed in a subgroup of 107 patients found negative for BRCA1 exon 11 mutations. CONCLUSIONS Our data suggest that BRCA1 methylation determines the earlier onset of HGSOC. Furthermore our study supports the idea that BRCAness is not only due to mutations but also to epigenetic changes in BRCA1 promoter gene.
Collapse
Affiliation(s)
- I Ruscito
- Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353 Berlin, Germany; Department of Gynecology, Obstetrics and Urology, Sapienza University of Rome, Rome, Italy
| | - D Dimitrova
- Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - I Vasconcelos
- Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - K Gellhaus
- Ivana Türbachova Laboratory for Epigenetics, Epiontis GMBH, Berlin, Germany
| | - T Schwachula
- Ivana Türbachova Laboratory for Epigenetics, Epiontis GMBH, Berlin, Germany
| | - F Bellati
- Department of Gynecology, Obstetrics and Urology, Sapienza University of Rome, Rome, Italy
| | - R Zeillinger
- Department of Obstetrics and Gynecology, Molecular Oncology Group, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | - P Benedetti-Panici
- Department of Gynecology, Obstetrics and Urology, Sapienza University of Rome, Rome, Italy
| | - I Vergote
- Division of Gynaecological Oncology, Department of Obstetrics and Gynaecology, Universitaire Ziekenhuizen Leuven, Katholieke Universiteit Leuven, UZ Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - S Mahner
- Department of Gynecology and Gynecologic Oncology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - D Cacsire-Tong
- Ivana Türbachova Laboratory for Epigenetics, Epiontis GMBH, Berlin, Germany
| | - N Concin
- Department of Gynecology and Obstetrics, Innsbruck Medical University, Anichstrasse 35, 6020 Innsbruck, Austria
| | - S Darb-Esfahani
- Institute of Pathology, Charite Medical University, Berlin, Campus Mitte, Germany
| | - S Lambrechts
- Division of Gynaecological Oncology, Department of Obstetrics and Gynaecology, Universitaire Ziekenhuizen Leuven, Katholieke Universiteit Leuven, UZ Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - J Sehouli
- Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - S Olek
- Ivana Türbachova Laboratory for Epigenetics, Epiontis GMBH, Berlin, Germany
| | - E I Braicu
- Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353 Berlin, Germany.
| |
Collapse
|
81
|
LAN VOTHITHUONG, HA NGOTHI, UYEN NGUYENQUYNH, DUONG NGUYENTHI, HUONG NGUYENTHITHU, THUAN TABICH, DUONG PHAMANHTHUY, VAN TO TA. Standardization of the methylation-specific PCR method for analyzing BRCA1 and ER methylation. Mol Med Rep 2014; 9:1844-50. [DOI: 10.3892/mmr.2014.1990] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 02/05/2014] [Indexed: 11/06/2022] Open
|
82
|
Cunningham JM, Cicek MS, Larson NB, Davila J, Wang C, Larson MC, Song H, Dicks EM, Harrington P, Wick M, Winterhoff BJ, Hamidi H, Konecny GE, Chien J, Bibikova M, Fan JB, Kalli KR, Lindor NM, Fridley BL, Pharoah PPD, Goode EL. Clinical characteristics of ovarian cancer classified by BRCA1, BRCA2, and RAD51C status. Sci Rep 2014; 4:4026. [PMID: 24504028 PMCID: PMC4168524 DOI: 10.1038/srep04026] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 01/20/2014] [Indexed: 12/20/2022] Open
Abstract
We evaluated homologous recombination deficient (HRD) phenotypes in epithelial ovarian cancer (EOC) considering BRCA1, BRCA2, and RAD51C in a large well-annotated patient set. We evaluated EOC patients for germline deleterious mutations (n = 899), somatic mutations (n = 279) and epigenetic alterations (n = 482) in these genes using NGS and genome-wide methylation arrays. Deleterious germline mutations were identified in 32 (3.6%) patients for BRCA1, in 28 (3.1%) for BRCA2 and in 26 (2.9%) for RAD51C. Ten somatically sequenced patients had deleterious alterations, six (2.1%) in BRCA1 and four (1.4%) in BRCA2. Fifty two patients (10.8%) had methylated BRCA1 or RAD51C. HRD patients with germline or somatic alterations in any gene were more likely to be high grade serous, have an earlier diagnosis age and have ovarian and/or breast cancer family history. The HRD phenotype was most common in high grade serous EOC. Identification of EOC patients with an HRD phenotype may help tailor specific therapies.
Collapse
Affiliation(s)
- J. M. Cunningham
- Department of Laboratory Medicine and Pathology, Division of Experimental Pathology, Mayo Clinic, Rochester, Minnesota
| | - M. S. Cicek
- Department of Health Sciences Research, Division of Epidemiology, Mayo Clinic, Rochester, Minnesota
| | - N. B. Larson
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - J. Davila
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - C. Wang
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - M. C. Larson
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - H. Song
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - E. M. Dicks
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - P. Harrington
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - M. Wick
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota
| | - B. J. Winterhoff
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota
| | - H. Hamidi
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California at Los Angeles and Jonsson Comprehensive Cancer Center, Los Angeles, California
| | - G. E. Konecny
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California at Los Angeles and Jonsson Comprehensive Cancer Center, Los Angeles, California
| | - J. Chien
- Department of Translational Genomics, University of Kansas Medical Center, Kansas City, Kansas
| | | | - J.-B. Fan
- Illumina Corporation, San Diego, California
| | - K. R. Kalli
- Department of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - N. M. Lindor
- Department of Health Science Research, Medical Genetics, Mayo Clinic, Scottsdale, Arizona
| | - B. L. Fridley
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, Kansas
| | - P. P. D. Pharoah
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - E. L. Goode
- Department of Health Sciences Research, Division of Epidemiology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
83
|
Yiannakopoulou E. Etiology of familial breast cancer with undetected BRCA1 and BRCA2 mutations: clinical implications. Cell Oncol (Dordr) 2013; 37:1-8. [PMID: 24306927 DOI: 10.1007/s13402-013-0158-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2013] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Familial breast cancer accounts for 20-30 % of all breast cancer cases. Mutations in the BRCA1 and BRCA2 genes account for the majority of high risk families with both early onset breast cancer and ovarian cancer. Most of the families with less than six breast cancer cases and no ovarian cancer do not carry BRCA1 or BRCA2 mutations that can be detected using routine sequencing protocols. Here, we aimed to review the etiology of familial breast cancer in cases without BRCA1 and BRCA2 mutations. RESULTS After excluding BRCA1 and BRCA2 mutations, factors proposed to contribute to familial breast cancer include: chance clustering of apparently sporadic cases, shared lifestyle, monogenic inheritance, i.e., dominant gene mutations associated with a high risk (TP53, PTEN, STK11), dominant gene mutations associated with a relatively low risk (ATM, BRIP1, RLB2), recessive gene mutations associated with horizontal inheritance patterns (sister-sister), and polygenic inheritance where susceptibility to familial breast cancer is thought to be conferred by a large number of low risk alleles. CONCLUSIONS Current evidence suggests that in the majority of cases with BRCA1 and BRCA2 negative familial breast cancer the etiology is due to interactions of intermediate or low risk alleles with environmental and lifestyle factors. Thus, a careful selection of patients submitted to genetic testing is needed. Clearly, further research is required to fully elucidate the etiology of non-BRCA familial breast cancer.
Collapse
Affiliation(s)
- Eugenia Yiannakopoulou
- Department of Basic Medical Lessons Faculty of Health and Caring Professions, Technological Educational Institute of Athens, Eleutheriou Benizelou 106 Kallithea, 17676, Athens, Greece,
| |
Collapse
|
84
|
Epimutations mimic genomic mutations of DNMT3A in acute myeloid leukemia. Leukemia 2013; 28:1227-34. [PMID: 24280869 PMCID: PMC4051212 DOI: 10.1038/leu.2013.362] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 11/20/2013] [Accepted: 11/22/2013] [Indexed: 12/29/2022]
Abstract
Mutations in the genetic sequence of the DNA de novo methyltransferase DNMT3A (DNA methyltransferase 3A) are found in many patients with acute myeloid leukemia (AML). They lead to dysfunction of DNMT3A protein and represent a marker for poor prognosis. Effects of genetic mutations can be mimicked by epigenetic modifications in the DNA methylation (DNAm) pattern. Using DNAm profiles of the Cancer Genome Atlas Research Network (TCGA), we identified aberrant hypermethylation at an internal promoter region of DNMT3A, which occurred in about 40% of AML patients. Bisulfite pyrosequencing assays designed for this genomic region validated hypermethylation specifically in a subset of our AML samples. High DNAm levels at this site are particularly observed in samples without genetic mutations in DNMT3A. Epimutations and mutations of DNMT3A were associated with related gene expression changes such as upregulation of the homeobox genes in HOXA and HOXB clusters. Furthermore, epimutations in DNMT3A were enriched in patients with poor or intermediate cytogenetic risk, and in patients with shorter event-free survival and overall survival (OS). Taken together, aberrant DNA hypermethylation within the DNMT3A gene, in analogy to DNMT3A mutations, is frequently observed in AML and both modifications seem to be useful for risk stratification or choice of therapeutic regimen.
Collapse
|
85
|
Lehnen H, Zechner U, Haaf T. Epigenetics of gestational diabetes mellitus and offspring health: the time for action is in early stages of life. Mol Hum Reprod 2013; 19:415-22. [PMID: 23515667 PMCID: PMC3690806 DOI: 10.1093/molehr/gat020] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 03/12/2013] [Accepted: 03/14/2013] [Indexed: 12/16/2022] Open
Abstract
The epidemic increase of type 2 diabetes and obesity in developed countries cannot be explained by overnutrition, physical inactivity and/or genetic factors alone. Epidemiologic evidence suggests that an adverse intrauterine environment, in particular a shortage or excess of nutrients is associated with increased risks for many complex diseases later in life. An impressive example for the 'fetal origins of adult disease' is gestational diabetes mellitus which usually presents in 1% to >10% of third trimester pregnancies. Intrauterine hyperglycemia is not only associated with increased perinatal morbidity and mortality, but also with increased lifelong risks of the exposed offspring for obesity, metabolic, cardiovascular and malignant diseases. Accumulating evidence suggests that fetal overnutrition (and similarly undernutrition) lead to persistent epigenetic changes in developmentally important genes, influencing neuroendocrine functions, energy homeostasis and metabolism. The concept of fetal programming has important implications for reproductive medicine. Because during early development the epigenome is much more vulnerable to environmental cues than later in life, avoiding adverse environmental factors in the periconceptional and intrauterine period may be much more important for the prevention of adult disease than any (i.e. dietetic) measures in infants and adults. A successful pregnancy should not primarily be defined by the outcome at birth but also by the health status in later life.
Collapse
Affiliation(s)
- Harald Lehnen
- Department of Gynecology and Obstetrics, Municipal Clinics, Hafenstrasse 100, 41239 Moenchengladbach, Germany
| | - Ulrich Zechner
- Institute of Human Genetics, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Thomas Haaf
- Institute of Human Genetics, Julius Maximilians University Wuerzburg, Biozentrum, Am Hubland, 97074 Wuerzburg, Germany
| |
Collapse
|
86
|
Basic concepts of epigenetics. Fertil Steril 2013; 99:607-15. [DOI: 10.1016/j.fertnstert.2013.01.117] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 01/16/2013] [Accepted: 01/17/2013] [Indexed: 12/20/2022]
|