1
|
Śniadecki M, Brzeziński M, Darecka K, Klasa-Mazurkiewicz D, Poniewierza P, Krzeszowiec M, Kmieć N, Wydra D. BARD1 and Breast Cancer: The Possibility of Creating Screening Tests and New Preventive and Therapeutic Pathways for Predisposed Women. Genes (Basel) 2020; 11:genes11111251. [PMID: 33114377 PMCID: PMC7693009 DOI: 10.3390/genes11111251] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 10/10/2020] [Indexed: 02/07/2023] Open
Abstract
Current oncological developments are based on improved understanding of genetics, and especially the discovery of genes whose alterations affect cell functions with consequences for the whole body. Our work is focused on the one of these genes, BRCA1-associated RING domain protein 1 (BARD1), and its oncogenic role in breast cancer. Most importantly, the study points to new avenues in the treatment and prevention of the most frequent female cancer based on BARD1 research. The BARD1 and BRCA1 (BReast CAncer type 1) proteins have similar structures and functions, and they combine to form the new molecule BARD1-BRCA1 heterodimer. The BARD1-BRCA1 complex is involved in genetic stabilization at the cellular level. It allows to mark abnormal DNA fragments by attaching ubiquitin to them. In addition, it blocks (by ubiquitination of RNA polymerase II) the transcription of damaged DNA. Ubiquitination, as well as stabilizing chromatin, or regulating the number of centrosomes, confirms the protective cooperation of BARD1 and BRCA1 in the stabilization of the genome. The overexpression of the oncogenic isoforms BARD1β and BARD1δ permit cancer development. The introduction of routine tests, for instance, to identify the presence of the BARD1β isoform, would make it possible to detect patients at high risk of developing cancer. On the other hand, introducing BARD1δ isoform blocking therapy, which would reduce estrogen sensitivity, may be a new line of cancer therapy with potential to modulate responses to existing treatments. It is possible that the BARD 1 gene offers new hope for improving breast cancer therapy.
Collapse
Affiliation(s)
- Marcin Śniadecki
- Department of Gynecology, Gynecologic Endocrinology and Gynecologic Oncology, Medical University of Gdańsk, Prof. Marian Smoluchowski Str. No. 17, 80-214 Gdańsk, Poland; (M.B.); (D.K.-M.); (M.K.); (D.W.)
- Correspondence: ; Tel.: +48-501-337-941
| | - Michał Brzeziński
- Department of Gynecology, Gynecologic Endocrinology and Gynecologic Oncology, Medical University of Gdańsk, Prof. Marian Smoluchowski Str. No. 17, 80-214 Gdańsk, Poland; (M.B.); (D.K.-M.); (M.K.); (D.W.)
| | - Katarzyna Darecka
- St. Adalbert’s Hospital, Department of Gynecology and Obstetrics, St. Jean Paul 2nd No. 50 Avenue, 80-462 Gdańsk, Poland;
| | - Dagmara Klasa-Mazurkiewicz
- Department of Gynecology, Gynecologic Endocrinology and Gynecologic Oncology, Medical University of Gdańsk, Prof. Marian Smoluchowski Str. No. 17, 80-214 Gdańsk, Poland; (M.B.); (D.K.-M.); (M.K.); (D.W.)
| | - Patryk Poniewierza
- Warsaw College of Engineering and Health, The Battle of Warsaw 1920. Str. No. 18, 02-366 Warsaw, Poland;
| | - Marta Krzeszowiec
- Department of Gynecology, Gynecologic Endocrinology and Gynecologic Oncology, Medical University of Gdańsk, Prof. Marian Smoluchowski Str. No. 17, 80-214 Gdańsk, Poland; (M.B.); (D.K.-M.); (M.K.); (D.W.)
| | - Natalia Kmieć
- Department of Oncology and Radiotherapy, University Clinical Center in Gdańsk, Prof. Marian Smoluchowski Str. No. 17, 80-214 Gdańsk, Poland;
| | - Dariusz Wydra
- Department of Gynecology, Gynecologic Endocrinology and Gynecologic Oncology, Medical University of Gdańsk, Prof. Marian Smoluchowski Str. No. 17, 80-214 Gdańsk, Poland; (M.B.); (D.K.-M.); (M.K.); (D.W.)
| |
Collapse
|
2
|
Cheng AC, Shen CJ, Hung CM, Hsu YC. Sulforaphane Decrease of SERTAD1 Expression Triggers G1/S Arrest in Breast Cancer Cells. J Med Food 2019; 22:444-450. [PMID: 31084542 PMCID: PMC6534085 DOI: 10.1089/jmf.2018.4195] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Studies have identified the potential of chemopreventive effects of sulforaphane (SFN); however, the underlying mechanisms of its effect on breast cancer require further elucidation. This study investigated the anticancer effects of SFN that specifically induces G1/S arrest in breast ductal carcinoma (ZR-75-1) cells. The proliferation of the cancer cells after treatment with SFN was detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. DNA content and cell cycle status were analyzed through flow cytometry. Our results demonstrated the inhibition of growth in ZR-75-1 cells upon SFN exposure. In addition, SERTAD1 (SEI-1) caused the accumulation of SFN-treated G1/S-phase cells. The downregulation of SEI-1, cyclin D2, and histone deacetylase 3 suggested that in addition to the identified effects of SFN against breast cancer prevention, it may also exert antitumor activities in established breast cancer cells. In conclusion, SFN can inhibit growth of and induce cell cycle arrest in cancer cells, suggesting its potential role as an anticancer agent.
Collapse
Affiliation(s)
- An-Chin Cheng
- 1 Department of Nutrition and Health Sciences; College of Health Sciences; Chang Jung Christian University, Tainan, Taiwan
| | - Ching-Ju Shen
- 2 Department of Gynecology and Obstetrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chao-Ming Hung
- 3 Department of General Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Yi-Chiang Hsu
- 4 Department of Medical Sciences Industry, College of Health Sciences; Chang Jung Christian University, Tainan, Taiwan
| |
Collapse
|
3
|
Felicio PS, Bidinotto LT, Melendez ME, Grasel RS, Campacci N, Galvão HCR, Scapulatempo-Neto C, Dufloth RM, Evangelista AF, Palmero EI. Genetic alterations detected by comparative genomic hybridization in BRCAX breast and ovarian cancers of Brazilian population. Oncotarget 2018; 9:27525-27534. [PMID: 29938003 PMCID: PMC6007956 DOI: 10.18632/oncotarget.25537] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 05/14/2018] [Indexed: 12/19/2022] Open
Abstract
Background About 5–10% of breast/ovarian cancers are hereditary. However, for a large proportion of cases (around 50%), the genetic cause remains unknown. These cases are grouped in a separated BRCAX category. The aim of this study was to identify genomic alterations in BRCA1/BRCA2 wild-type tumor samples from women with family history strongly suggestive of hereditary breast/ovarian cancer. Results A cohort of 31 Brazilian women was included in the study. Using the GISTIC algorithm, we identified 20 regions with genomic gains and 31 with losses. The most frequent altered regions were 1q21.2, 6p22.1 and 8p23.3 in breast tumors and Xq26 and Xp22.32-22.31 among the ovarian cancer cases. An interesting association identified was the loss of 22q13.31-13.32 and the presence of ovarian cancer cases. Among the genes present in the frequently altered regions, we found FGFR1, NSMCE2, CTTN, CRLF2, ERBB2, STARD3, MIR3201 and several genes of RAET and ULBP family. Conclusions In conclusion, our results suggest that alterations on chromosomes 1, 6, 8 and X are common on BRCAX tumors and that the loss on 22q can be associated with the presence of ovarian cancer. Methods DNA copy number alterations were analyzed by 60K array comparative genomic hybridization in breast and ovarian FFPE tumors.
Collapse
Affiliation(s)
- Paula Silva Felicio
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, SP, Brazil
| | - Lucas Tadeu Bidinotto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, SP, Brazil.,Barretos School of Health Sciences-FACISB, Barretos, SP, Brazil
| | | | | | - Natalia Campacci
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, SP, Brazil
| | | | | | | | | | - Edenir Inêz Palmero
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, SP, Brazil.,Barretos School of Health Sciences-FACISB, Barretos, SP, Brazil
| |
Collapse
|
4
|
Johnson J, Bessette DC, Saunus JM, Smart CE, Song S, Johnston RL, Cocciardi S, Rozali EN, Johnstone CN, Vargas AC, Kazakoff SH, BioBank VC, Khanna KK, Lakhani SR, Chenevix-Trench G, Simpson PT, Nones K, Waddell N, Al-Ejeh F. Characterization of a novel breast cancer cell line derived from a metastatic bone lesion of a breast cancer patient. Breast Cancer Res Treat 2018; 170:179-188. [DOI: 10.1007/s10549-018-4719-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 02/15/2018] [Indexed: 02/03/2023]
|
5
|
Bao Y, Liu J, You J, Wu D, Yu Y, Liu C, Wang L, Wang F, Xu L, Wang L, Wang N, Tian X, Wang F, Liang H, Gao Y, Cui X, Ji G, Bai J, Yu J, Meng X, Jin Y, Sun W, Guan XY, Zhang C, Fu S. Met promotes the formation of double minute chromosomes induced by Sei-1 in NIH-3T3 murine fibroblasts. Oncotarget 2018; 7:56664-56675. [PMID: 27494853 PMCID: PMC5302943 DOI: 10.18632/oncotarget.10994] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/19/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Sei-1 is an oncogene capable of inducing double minute chromosomes (DMs) formation. DMs are hallmarks of amplification and contribute to oncogenesis. However, the mechanism of Sei-1 inducing DMs formation remains unelucidated. RESULTS DMs formation significantly increased during serial passage in vivo and gradually decreased following culture in vitro. micro nuclei (MN) was found to be responsible for the reduction. Of the DMs-carrying genes, Met was found to be markedly amplified, overexpressed and highly correlated with DMs formation. Inhibition of Met signaling decreased the number of DMs and reduced the amplification of the DMs-carrying genes. We identified a 3.57Mb DMs representing the majority population, which consists of the 1.21 Mb AMP1 from locus 6qA2 and the 2.36 Mb AMP2 from locus 6qA2-3. MATERIALS AND METHODS We employed NIH-3T3 cell line with Sei-1 overexpression to monitor and characterize DMs in vivo and in vitro. Array comparative genome hybridization (aCGH) and fluorescence in situ hybridization (FISH) were performed to reveal amplification regions and DMs-carrying genes. Metaphase spread was prepared to count the DMs. Western blot and Met inhibition rescue experiments were performed to examine for involvement of altered Met signaling in Sei-1 induced DMs. Genomic walking and PCR were adopted to reveal DMs structure. CONCLUSIONS Met is an important promotor of DMs formation.
Collapse
Affiliation(s)
- Yantao Bao
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Jia Liu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Jia You
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Di Wu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Yang Yu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China.,Department of Genetics and Eugenics, Maternity and Child Care Center of Qinghuangdao, Qinghuangdao, China
| | - Chang Liu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Lei Wang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China.,Genetic Diagnosis Center, First People's Hospital of Yunnan Province, Yunnan, China
| | - Fei Wang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Lu Xu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Liqun Wang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Nan Wang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Xing Tian
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Falin Wang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Hongbin Liang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Yating Gao
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Xiaobo Cui
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Guohua Ji
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Jing Bai
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Jingcui Yu
- Scientific Research Centre, Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xiangning Meng
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Yan Jin
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Wenjing Sun
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chunyu Zhang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Songbin Fu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China.,Key Laboratory of Medical Genetics, Harbin Medical University, Heilongjiang Higher Education Institutions, Harbin, China
| |
Collapse
|
6
|
Non-BRCA familial breast cancer: review of reported pathology and molecular findings. Pathology 2017; 49:363-370. [PMID: 28450088 DOI: 10.1016/j.pathol.2017.03.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/07/2017] [Accepted: 03/16/2017] [Indexed: 01/02/2023]
Abstract
The majority of women evaluated for a clinical concern of possible hereditary breast cancer syndromes have no identified pathogenic variants in genes predisposing them to breast cancer. Non-BRCA1- or BRCA2-related familial breast cancer, also called 'BRCAX', thus comprises a sizeable proportion of familial breast cancer but it is poorly understood. In this study, we reviewed 14 studies on histopathology and molecular studies of BRCAX to determine if there were differences between 'sporadic' breast cancers and compared to cancers arising in women harbouring variants in known cancer predisposition genes. Across available literature, there was inconsistency on inclusion and exclusion criteria, reported parameters, and use of controls. Cohorts were small, and while several studies reported findings that appeared to distinguish the BRCAX cases from sporadic and/or gene-positive controls, no findings were reported in more than one study. To determine whether the BRCAX families might still contain important genetic subsets awaiting discovery will require prospective ascertainment of a large number of women with familial breast cancer who are screened for all currently established predisposition genes, whose tumours are assessed for multiple parameters in a uniform manner, and in which controls (BRCA1/2+ and non-familial 'sporadic' cases) are collected from the same population.
Collapse
|
7
|
Alvarez C, Aravena A, Tapia T, Rozenblum E, Solís L, Corvalán A, Camus M, Alvarez M, Munroe D, Maass A, Carvallo P. Different Array CGH profiles within hereditary breast cancer tumors associated to BRCA1 expression and overall survival. BMC Cancer 2016; 16:219. [PMID: 26979459 PMCID: PMC4791866 DOI: 10.1186/s12885-016-2261-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 03/08/2016] [Indexed: 12/20/2022] Open
Abstract
Background Array CGH analysis of breast tumors has contributed to the identification of different genomic profiles in these tumors. Loss of DNA repair by BRCA1 functional deficiency in breast cancer has been proposed as a relevant contribution to breast cancer progression for tumors with no germline mutation. Identifying the genomic alterations taking place in BRCA1 not expressing tumors will lead us to a better understanding of the cellular functions affected in this heterogeneous disease. Moreover, specific genomic alterations may contribute to the identification of potential therapeutic targets and offer a more personalized treatment to breast cancer patients. Methods Forty seven tumors from hereditary breast cancer cases, previously analyzed for BRCA1 expression, and screened for germline BRCA1 and 2 mutations, were analyzed by Array based Comparative Genomic Hybridization (aCGH) using Agilent 4x44K arrays. Overall survival was established for tumors in different clusters using Log-rank (Mantel-Cox) Test. Gene lists obtained from aCGH analysis were analyzed for Gene Ontology enrichment using GOrilla and DAVID tools. Results Genomic profiling of the tumors showed specific alterations associated to BRCA1 or 2 mutation status, and BRCA1 expression in the tumors, affecting relevant cellular processes. Similar cellular functions were found affected in BRCA1 not expressing and BRCA1 or 2 mutated tumors. Hierarchical clustering classified hereditary breast tumors in four major, groups according to the type and amount of genomic alterations, showing one group with a significantly poor overall survival (p = 0.0221). Within this cluster, deletion of PLEKHO1, GDF11, DARC, DAG1 and CD63 may be associated to the worse outcome of the patients. Conclusions These results support the fact that BRCA1 lack of expression in tumors should be used as a marker for BRCAness and to select these patients for synthetic lethality approaches such as treatment with PARP inhibitors. In addition, the identification of specific alterations in breast tumors associated with poor survival, immune response or with a BRCAness phenotype will allow the use of a more personalized treatment in these patients.
Collapse
Affiliation(s)
- Carolina Alvarez
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Andrés Aravena
- Mathomics, Center for Mathematical Modeling (UMI 2807 CNRS) and Center for Genome Regulation (Fondap 15090007), University of Chile, Santiago, Chile.,Department of Molecular Biology and Genetics, Faculty of Science, Istanbul University, Istanbul, 34134, Turkey
| | - Teresa Tapia
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ester Rozenblum
- Laboratory of Molecular Technology Advanced Technology Program, SAIC-Frederick, Inc., National Cancer Institute-Frederick, Frederick, MD, USA
| | - Luisa Solís
- Department of Anatomo-Pathology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandro Corvalán
- Department of Anatomo-Pathology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mauricio Camus
- Cancer Center, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - David Munroe
- Laboratory of Molecular Technology Advanced Technology Program, SAIC-Frederick, Inc., National Cancer Institute-Frederick, Frederick, MD, USA
| | - Alejandro Maass
- Mathomics, Center for Mathematical Modeling (UMI 2807 CNRS) and Center for Genome Regulation (Fondap 15090007), University of Chile, Santiago, Chile.,Department of Mathematical Engineering, University of Chile, Santiago, Chile
| | - Pilar Carvallo
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
8
|
Cassidy MR, Méndez JE. BRCA1 and BRCA2 in Breast Cancer and Ovarian Cancer. Surg Oncol 2015. [DOI: 10.1007/978-1-4939-1423-4_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
9
|
Muthuswami M, Ramesh V, Banerjee S, Viveka Thangaraj S, Periasamy J, Bhaskar Rao D, Barnabas GD, Raghavan S, Ganesan K. Breast tumors with elevated expression of 1q candidate genes confer poor clinical outcome and sensitivity to Ras/PI3K inhibition. PLoS One 2013; 8:e77553. [PMID: 24147022 PMCID: PMC3798322 DOI: 10.1371/journal.pone.0077553] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 09/03/2013] [Indexed: 12/15/2022] Open
Abstract
Genomic aberrations are common in cancers and the long arm of chromosome 1 is known for its frequent amplifications in breast cancer. However, the key candidate genes of 1q, and their contribution in breast cancer pathogenesis remain unexplored. We have analyzed the gene expression profiles of 1635 breast tumor samples using meta-analysis based approach and identified clinically significant candidates from chromosome 1q. Seven candidate genes including exonuclease 1 (EXO1) are consistently over expressed in breast tumors, specifically in high grade and aggressive breast tumors with poor clinical outcome. We derived a EXO1 co-expression module from the mRNA profiles of breast tumors which comprises 1q candidate genes and their co-expressed genes. By integrative functional genomics investigation, we identified the involvement of EGFR, RAS, PI3K / AKT, MYC, E2F signaling in the regulation of these selected 1q genes in breast tumors and breast cancer cell lines. Expression of EXO1 module was found as indicative of elevated cell proliferation, genomic instability, activated RAS/AKT/MYC/E2F1 signaling pathways and loss of p53 activity in breast tumors. mRNA-drug connectivity analysis indicates inhibition of RAS/PI3K as a possible targeted therapeutic approach for the patients with activated EXO1 module in breast tumors. Thus, we identified seven 1q candidate genes strongly associated with the poor survival of breast cancer patients and identified the possibility of targeting them with EGFR/RAS/PI3K inhibitors.
Collapse
Affiliation(s)
- Muthulakshmi Muthuswami
- Cancer Genetics Laboratory, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Vignesh Ramesh
- Cancer Genetics Laboratory, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Saikat Banerjee
- Cancer Genetics Laboratory, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Soundara Viveka Thangaraj
- Cancer Genetics Laboratory, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Jayaprakash Periasamy
- Cancer Genetics Laboratory, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Divya Bhaskar Rao
- Cancer Genetics Laboratory, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Georgina D. Barnabas
- Cancer Genetics Laboratory, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Swetha Raghavan
- Department of Biotechnology, Indian Institute of Technology Madras, Chenna, India
| | - Kumaresan Ganesan
- Cancer Genetics Laboratory, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| |
Collapse
|
10
|
The complex genetic landscape of familial breast cancer. Hum Genet 2013; 132:845-63. [PMID: 23552954 DOI: 10.1007/s00439-013-1299-y] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 03/21/2013] [Indexed: 01/19/2023]
Abstract
Familial breast cancer represents a minor percentage of all human breast cancers. Mutations in two high susceptibility genes BRCA1 and BRCA2 explain around 25 % of familial breast cancers, while other high, moderate and low susceptibility genes explain up to 20 % more of breast cancer families. Thus, it is important to decipher the genetic architecture of families that show no mutations to improve genetic counselling. The comprehensive description of familial breast cancer using different techniques and platforms has shown to be very valuable for better patient diagnosis, tumour surveillance, and ultimately patient treatment. This review focuses on the complex landscape of pathological, protein, genetic and genomic features associated with BRCA1-, BRCA2-, and non-BRCA1/BRCA2-related cancers described up to date. Special emphasis deserves the coexistence of distinct molecular breast cancer subtypes, the development of tumour classifiers to predict BRCA1/2 mutations, and the last insights from recent whole genome sequencing studies and miRNA profiling.
Collapse
|
11
|
DNA copy number profiling reveals extensive genomic loss in hereditary BRCA1 and BRCA2 ovarian carcinomas. Br J Cancer 2013; 108:1732-42. [PMID: 23558894 PMCID: PMC3668473 DOI: 10.1038/bjc.2013.141] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: Few studies have attempted to characterise genomic changes occurring in hereditary epithelial ovarian carcinomas (EOCs) and inconsistent results have been obtained. Given the relevance of DNA copy number alterations in ovarian oncogenesis and growing clinical implications of the BRCA-gene status, we aimed to characterise the genomic profiles of hereditary and sporadic ovarian tumours. Methods: High-resolution array Comparative Genomic Hybridisation profiling of 53 familial (21 BRCA1, 6 BRCA2 and 26 non-BRCA1/2) and 15 sporadic tumours in combination with supervised and unsupervised analysis was used to define common and/or specific copy number features. Results: Unsupervised hierarchical clustering did not stratify tumours according to their familial or sporadic condition or to their BRCA1/2 mutation status. Common recurrent changes, spanning genes potentially fundamental for ovarian carcinogenesis, regardless of BRCA mutations, and several candidate subtype-specific events were defined. Despite similarities, greater contribution of losses was revealed to be a hallmark of BRCA1 and BRCA2 tumours. Conclusion: Somatic alterations occurring in the development of familial EOCs do not differ substantially from the ones occurring in sporadic carcinomas. However, some specific features like extensive genomic loss observed in BRCA1/2 tumours may be of clinical relevance helping to identify BRCA-related patients likely to respond to PARP inhibitors.
Collapse
|
12
|
Hilbers FS, Meijers CM, Laros JFJ, van Galen M, Hoogerbrugge N, Vasen HFA, Nederlof PM, Wijnen JT, van Asperen CJ, Devilee P. Exome sequencing of germline DNA from non-BRCA1/2 familial breast cancer cases selected on the basis of aCGH tumor profiling. PLoS One 2013; 8:e55734. [PMID: 23383274 PMCID: PMC3561352 DOI: 10.1371/journal.pone.0055734] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 12/30/2012] [Indexed: 12/17/2022] Open
Abstract
The bulk of familial breast cancer risk (∼70%) cannot be explained by mutations in the known predisposition genes, primarily BRCA1 and BRCA2. Underlying genetic heterogeneity in these cases is the probable explanation for the failure of all attempts to identify further high-risk alleles. While exome sequencing of non-BRCA1/2 breast cancer cases is a promising strategy to detect new high-risk genes, rational approaches to the rigorous pre-selection of cases are needed to reduce heterogeneity. We selected six families in which the tumours of multiple cases showed a specific genomic profile on array comparative genomic hybridization (aCGH). Linkage analysis in these families revealed a region on chromosome 4 with a LOD score of 2.49 under homogeneity. We then analysed the germline DNA of two patients from each family using exome sequencing. Initially focusing on the linkage region, no potentially pathogenic variants could be identified in more than one family. Variants outside the linkage region were then analysed, and we detected multiple possibly pathogenic variants in genes that encode DNA integrity maintenance proteins. However, further analysis led to the rejection of all variants due to poor co-segregation or a relatively high allele frequency in a control population. We concluded that using CGH results to focus on a sub-set of families for sequencing analysis did not enable us to identify a common genetic change responsible for the aggregation of breast cancer in these families. Our data also support the emerging view that non-BRCA1/2 hereditary breast cancer families have a very heterogeneous genetic basis.
Collapse
Affiliation(s)
- Florentine S Hilbers
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Saponaro C, Malfettone A, Ranieri G, Danza K, Simone G, Paradiso A, Mangia A. VEGF, HIF-1α expression and MVD as an angiogenic network in familial breast cancer. PLoS One 2013; 8:e53070. [PMID: 23326384 PMCID: PMC3543407 DOI: 10.1371/journal.pone.0053070] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 11/28/2012] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis, which plays an important role in tumor growth and progression of breast cancer, is regulated by a balance between pro- and anti-angiogenic factors. Expression of vascular endothelial growth factor (VEGF) is up-regulated during hypoxia by hypoxia-inducible factor-1α (HIF-1α). It is known that there is an interaction between HIF-1α and BRCA1 carrier cancers, but little has been reported about angiogenesis in BRCA1-2 carrier and BRCAX breast cancers. In this study, we investigated the expression of VEGF and HIF-1α and microvessel density (MVD) in 26 BRCA1-2 carriers and 58 BRCAX compared to 77 sporadic breast cancers, by immunohistochemistry. VEGF expression in BRCA1-2 carriers was higher than in BRCAX cancer tissues (p = 0.0001). Furthermore, VEGF expression was higher in both BRCA1-2 carriers and BRCAX than the sporadic group (p<0.0001). VEGF immunoreactivity was correlated with poor tumor grade (p = 0.0074), hormone receptors negativity (p = 0.0206, p = 0.0002 respectively), and MIB-1-labeling index (p = 0.0044) in familial cancers (BRCA1-2 and BRCAX). The percentage of nuclear HIF-1α expression was higher in the BRCA1-2 carriers than in BRCAX cancers (p<0.05), and in all familial than in sporadic tumor tissues (p = 0.0045). A higher MVD was observed in BRCA1-2 carrier than in BRCAX and sporadic cancer tissues (p = 0.002, p = 0.0001 respectively), and in all familial tumors than in sporadic tumors (p = 0.01). MVD was positively related to HIF-1α expression in BRCA1-2 carriers (r = 0.521, p = 0.006), and, in particular, we observed a highly significant correlation in the familial group (r = 0.421, p<0.0001). Our findings suggest that angiogenesis plays a crucial role in BRCA1-2 carrier breast cancers. Prospective studies in larger BRCA1-2 carrier series are needed to improve the best therapeutic strategies for this subgroup of breast cancer patients.
Collapse
Affiliation(s)
- Concetta Saponaro
- Functional Biomorphology Laboratory, National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Andrea Malfettone
- Functional Biomorphology Laboratory, National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Girolamo Ranieri
- Unit of Interventional Radiology, National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Katia Danza
- Molecular Genetics Laboratory, National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Giovanni Simone
- Pathology Department, National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Angelo Paradiso
- Scientific Direction, National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Anita Mangia
- Functional Biomorphology Laboratory, National Cancer Research Centre, Istituto Tumori “Giovanni Paolo II”, Bari, Italy
- * E-mail:
| |
Collapse
|
14
|
Sabatier R, Adélaïde J, Finetti P, Ferrari A, Huiart L, Sobol H, Chaffanet M, Birnbaum D, Bertucci F. BARD1 homozygous deletion, a possible alternative to BRCA1 mutation in basal breast cancer. Genes Chromosomes Cancer 2011; 49:1143-51. [PMID: 20842729 DOI: 10.1002/gcc.20822] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Hereditary breast cancers (BCs) are incompletely explained by BRCA genes abnormalities, as ∼70% of them are not associated with known genetic alterations. Array-based comparative genomic hybridization (aCGH) of tumors provides an opportunity for identifying new BC susceptibility genes. By analyzing our database of high-resolution aCGH profiles of 330 BCs, we identified a case with homozygous deletion of the entire BARD1 gene. The BARD1-deleted case displayed a familial history of BC and other clinico-pathological features of BRCAness, and a 17% probability of BRCA1/2 mutation. Analysis of constitutional DNA showed a BARD1 germline heterozygous deletion without BRCA1/2 mutation. Gene expression analysis using DNA microarrays classified the tumor as basal-like, with very low BARD1 and ID4 expression, but high expression of BRCA1, RAD51, PARP1, CHEK1, and FANCA. The tumor displayed a BRCA1-mutated expression profile. This is the first report of a non-BRCA1/2-mutated BC with somatic homozygous and germ-line heterozygous deletion of the entire BARD1 gene. This observation suggests that BARD1 might be a BC susceptibility gene that follows the Knudson rule. Identification of BARD1 deletion could have clinical applications including screening for hereditary forms. © 2010 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Renaud Sabatier
- Department of Molecular Oncology, Centre de Recherche en Cancérologie de Marseille, INSERM/UMR891, Marseille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Didraga MA, van Beers EH, Joosse SA, Brandwijk KIM, Oldenburg RA, Wessels LFA, Hogervorst FBL, Ligtenberg MJ, Hoogerbrugge N, Verhoef S, Devilee P, Nederlof PM. A non-BRCA1/2 hereditary breast cancer sub-group defined by aCGH profiling of genetically related patients. Breast Cancer Res Treat 2011; 130:425-36. [PMID: 21286804 DOI: 10.1007/s10549-011-1357-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Accepted: 01/17/2011] [Indexed: 02/04/2023]
Abstract
Germline mutations in BRCA1 and BRCA2 explain approximately 25% of all familial breast cancers. Despite intense efforts to find additional high-risk breast cancer genes (BRCAx) using linkage analysis, none have been reported thus far. Here we explore the hypothesis that BRCAx breast tumors from genetically related patients share a somatic genetic etiology that might be revealed by array comparative genomic hybridization (aCGH) profiling. As BRCA1 and BRCA2 tumors can be identified on the basis of specific genomic profiles, the same may be true for a subset of BRCAx families. Analyses used aCGH to compare 58 non-BRCA1/2 familial breast tumors (designated BRCAx) to sporadic (non-familiar) controls, BRCA1 and BRCA2 tumors. The selection criteria for BRCAx families included at least three cases of breast cancer diagnosed before the age of 60 in the family, and the absence of ovarian or male breast cancer. Hierarchical cluster analysis was performed to determine sub-groups within the BRCAx tumor class and family heterogeneity. Analysis of aCGH profiles of BRCAx tumors indicated that they constitute a heterogeneous class, but are distinct from both sporadic and BRCA1/2 tumors. The BRCAx class could be divided into sub-groups. One subgroup was characterized by a gain of chromosome 22. Tumors from family members were classified within the same sub-group in agreement with the hypothesis that tumors from the same family would harbor a similar genetic background. This approach provides a method to target a sub-group of BRCAx families for further linkage analysis studies.
Collapse
Affiliation(s)
- M A Didraga
- Department of Experimental Therapy, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Fernández-Ramires R, Gómez G, Muñoz-Repeto I, de Cecco L, Llort G, Cazorla A, Blanco I, Gariboldi M, Pierotti MA, Benítez J, Osorio A. Transcriptional characteristics of familial non-BRCA1/BRCA2 breast tumors. Int J Cancer 2010; 128:2635-44. [PMID: 20715112 DOI: 10.1002/ijc.25603] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 07/23/2010] [Indexed: 12/12/2022]
Abstract
To better understand the alterations present in the group of the so-called BRCAX tumors, we have used a cDNA microarray containing genes related to tumorigenesis and analyzed a series of 49 tumors consisting of 13 BRCA1, 14 BRCAX and 22 sporadic. We have confirmed that the BRCAX tumors are heterogeneous and can be divided in at least two main subgroups, so-called A and B, transcriptionally distinguishable and with different altered pathways within each of the groups. We have found that BRCAX-A and B subgroups, can be classified as Luminal A and Luminal B, respectively, taking into account the intrinsic phenotypes defined for the sporadic breast tumors. We have found that, at the somatic level, the BRCAX-B tumors are identical to their sporadic Luminal B counterparts, whereas BRCAX-A, despite having a Luminal A phenotype, shows additional genomic alterations. We have found 21 deregulated genes in the BRCAX-A group that we have called "the BRCAX susceptibility pathway" and suggested it as a candidate to search for new genes involved in the inherited susceptibility underlying the disease in this group.
Collapse
Affiliation(s)
- Ricardo Fernández-Ramires
- Human Genetics Group, Human Cancer Genetics Program, Spanish National Cancer Center (CNIO) and CIBERER, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
de Plater L, Laugé A, Guyader C, Poupon MF, Assayag F, de Cremoux P, Vincent-Salomon A, Stoppa-Lyonnet D, Sigal-Zafrani B, Fontaine JJ, Brough R, Lord CJ, Ashworth A, Cottu P, Decaudin D, Marangoni E. Establishment and characterisation of a new breast cancer xenograft obtained from a woman carrying a germline BRCA2 mutation. Br J Cancer 2010; 103:1192-200. [PMID: 20877358 PMCID: PMC2967069 DOI: 10.1038/sj.bjc.6605900] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Background: The BRCA2 gene is responsible for a high number of hereditary breast and ovarian cancers, and studies of the BRCA2 biological functions are limited by the lack of models that resemble the patient's tumour features. The aim of this study was to establish and characterise a new human breast carcinoma xenograft obtained from a woman carrying a germline BRCA2 mutation. Methods: A transplantable xenograft was obtained by grafting a breast cancer sample into nude mice. The biological and genetic profiles of the xenograft were compared with that of the patient's tumour using histology, immunohistochemistry (IHC), BRCA2 sequencing, comparative genomic hybridisation (CGH), and qRT–PCR. Tumour response to standard chemotherapies was evaluated. Results: Histological profile identified the tumour as a basal-like triple-negative breast cancer. Targeted BRCA2 DNA sequencing of the xenograft showed the presence of the mutation previously identified in the carrier. Comparative genomic hybridisation array profiles of the primary tumour and the xenograft revealed a high number of similar genetic alterations. The therapeutic assessment of the xenograft showed sensitivity to anthracyclin-based chemotherapy and resistance to docetaxel. The xenograft was also highly sensitive to radiotherapy and cisplatin-based treatments. Conclusions: This study describes a new human breast cancer xenograft obtained from a BRCA2-mutated patient. This xenograft provides a new model for the pre-clinical drug development and for the exploration of the drug response biological basis.
Collapse
Affiliation(s)
- L de Plater
- Preclinical Investigation Unit, Institut Curie - Translational Research Department, Hôpital St Louis, Quadrilatère historique, Porte 13, 1, Ave Claude Vellefaux, Paris 75010, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Dong XY, Sun X, Guo P, Li Q, Sasahara M, Ishii Y, Dong JT. ATBF1 inhibits estrogen receptor (ER) function by selectively competing with AIB1 for binding to the ER in ER-positive breast cancer cells. J Biol Chem 2010; 285:32801-32809. [PMID: 20720010 DOI: 10.1074/jbc.m110.128330] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Loss of the q22 band of chromosome 16 is a frequent genetic event in breast cancer, and the candidate tumor suppressor gene, ATBF1, has been implicated in breast cancer by genomic deletion, transcriptional down-regulation, and association with better prognostic parameters. In addition, estrogen receptor (ER)-positive breast cancer expresses a higher level of ATBF1, suggesting a role of ATBF1 in ER-positive breast cancer. In this study, we examined whether and how ATBF1 affects the ER function in breast cancer cells. We found that ATBF1 inhibited ER-mediated gene transcription, cell growth, and proliferation in ER-positive breast cancer cells. In vitro and in vivo immunoprecipitation experiments revealed that ATBF1 interacted physically with the ER and that multiple domains in both ATBF1 and ER proteins mediated the interaction. Furthermore, we demonstrated that ATBF1 inhibited ER function by selectively competing with the steroid receptor coactivator AIB1 but not GRIP1 or SRC1 for binding to the ER. These findings not only support the concept that ATBF1 plays a tumor-suppressive role in breast cancer, they also provide a mechanism for how ATBF1 functions as a tumor suppressor in breast cancer.
Collapse
Affiliation(s)
- Xue-Yuan Dong
- From the Winship Cancer Institute and Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Xiaodong Sun
- From the Winship Cancer Institute and Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Peng Guo
- From the Winship Cancer Institute and Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Qunna Li
- From the Winship Cancer Institute and Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Masakiyo Sasahara
- Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, Toyama 930-0194, Japan
| | - Yoko Ishii
- Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, Toyama 930-0194, Japan
| | - Jin-Tang Dong
- From the Winship Cancer Institute and Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia 30322.
| |
Collapse
|
19
|
Li Y, Nie CJ, Hu L, Qin Y, Liu HB, Zeng TT, Chen L, Fu L, Deng W, Chen SP, Jia WH, Zhang C, Xie D, Guan XY. Characterization of a novel mechanism of genomic instability involving the SEI1/SET/NM23H1 pathway in esophageal cancers. Cancer Res 2010; 70:5695-705. [PMID: 20570897 DOI: 10.1158/0008-5472.can-10-0392] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Amplification of 19q is a frequent genetic alteration in many solid tumors, and SEI1 is a candidate oncogene within the amplified region. Our previous study found that the oncogenic function of SEI1 was associated with chromosome instability. In this study, we report a novel mechanism of genomic instability involving the SEI1-SET-NM23H1 pathway. Overexpression of SEI1 was observed in 57 of 100 of esophageal squamous cell carcinoma cases. Functional study showed that SEI1 had strong tumorigenic ability, and overexpression of SEI1 could induce the genomic instability by increasing micronuclei formation and reducing the number of chromosomes. Further study found that SEI1 was able to upregulate SET expression and subsequently promote the translocation of a small amount of NM23H1 from the cytoplasm to the nucleus. Nuclear NM23H1 can induce DNA damage through its DNA nick activity. Unlike CTL attack, only a small amount of NM23H1 translocated into the nucleus (<10%) induced by the overexpression of SEI1. Further study found that the small amount of NM23H1 only induced minor DNA damage and subsequently increased genomic instability, rather than inducing irreparable DNA damage and initiating apoptosis by CTL attack. Sister chromatid exchange experiment found that the translocation of small amount of NM23H1 into the nucleus induced by the overexpressions of SEI1/SET could increase the frequency of sister chromatid exchange. In addition, overexpression of SEI1 was associated with poor prognosis of esophageal squamous cell carcinoma. Taken together, these findings define a novel mechanism of genomic instability and malignant progression in esophageal cancers, a deadly disease of increasing incidence in developed countries.
Collapse
Affiliation(s)
- Yan Li
- State Key Laboratory of Oncology in Southern China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Waddell N, Arnold J, Cocciardi S, da Silva L, Marsh A, Riley J, Johnstone CN, Orloff M, Assie G, Eng C, Reid L, Keith P, Yan M, Fox S, Devilee P, Godwin AK, Hogervorst FBL, Couch F, Grimmond S, Flanagan JM, Khanna K, Simpson PT, Lakhani SR, Chenevix-Trench G. Subtypes of familial breast tumours revealed by expression and copy number profiling. Breast Cancer Res Treat 2009; 123:661-77. [PMID: 19960244 DOI: 10.1007/s10549-009-0653-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Accepted: 11/13/2009] [Indexed: 01/03/2023]
Abstract
Extensive expression profiling studies have shown that sporadic breast cancer is composed of five clinically relevant molecular subtypes. However, although BRCA1-related tumours are known to be predominantly basal-like, there are few published data on other classes of familial breast tumours. We analysed a cohort of 75 BRCA1, BRCA2 and non-BRCA1/2 breast tumours by gene expression profiling and found that 74% BRCA1 tumours were basal-like, 73% of BRCA2 tumours were luminal A or B, and 52% non-BRCA1/2 tumours were luminal A. Thirty-four tumours were also analysed by single nucleotide polymorphism-comparative genomic hybridization (SNP-CGH) arrays. Copy number data could predict whether a tumour was basal-like or luminal with high accuracy, but could not predict its mutation class. Basal-like BRCA1 and basal-like non-BRCA1 tumours were very similar, and contained the highest number of chromosome aberrations. We identified regions of frequent gain containing potential driver genes in the basal (8q and 12p) and luminal A tumours (1q and 17q). Regions of homozygous loss associated with decreased expression of potential tumour suppressor genes were also detected, including in basal tumours (5q and 9p), and basal and luminal tumours (10q). This study highlights the heterogeneity of familial tumours and the clinical consequences for treatment and prognosis.
Collapse
Affiliation(s)
- Nic Waddell
- Queensland Institute of Medical Research, Brisbane, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Oldenburg RA, Kroeze-Jansema KHG, Houwing-Duistermaat JJ, Bayley JP, Dambrot C, van Asperen CJ, van den Ouweland AMW, Bakker B, van Beers EH, Nederlof PM, Vasen H, Hoogerbrugge N, Cornelisse CJ, Meijers-Heijboer H, Devilee P. Genome-wide linkage scan in Dutch hereditary non-BRCA1/2 breast cancer families identifies 9q21-22 as a putative breast cancer susceptibility locus. Genes Chromosomes Cancer 2008; 47:947-56. [PMID: 18663745 DOI: 10.1002/gcc.20597] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Breast cancer accounts for over 20% of all female cancers. A positive family history remains one of the most important risk factors for the disease, with first-degree relatives of patients having a twofold elevated risk. Known breast cancer susceptibility genes such as BRCA1 and BRCA2 explain only 20-25% of this risk, suggesting the existence of other breast cancer susceptibility genes. Here, we report the results of a genome-wide linkage scan in 55 high-risk Dutch breast cancer families with no mutations in BRCA1 and BRCA2. Twenty-two of these families were also part of a previous linkage study by the Breast Cancer Linkage Consortium. In addition, we performed CGH analyses in 61 tumors of these families and 31 sporadic tumors. Three regions were identified with parametric HLOD scores >1, and three with nonparametric LOD scores >1.5. Upon further marker genotyping for the candidate loci, and the addition of another 30 families to the analysis, only the locus on chromosome 9 (9q21-22, marker D9S167) remained significant, with a nonparametric multipoint LOD score of 3.96 (parametric HLOD 0.56, alpha = 0.18). With CGH analyses we observed preferential copy number loss at BAC RP11-276H19, containing D9S167 in familial tumors as compared to sporadic tumors (P < 0.001). Five candidate genes were selected from the region around D9S167 and their coding regions subjected to direct sequence analysis in 16 probands. No clear pathogenic mutations were found in any of these genes.
Collapse
Affiliation(s)
- Rogier A Oldenburg
- Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Mangia A, Chiarappa P, Tommasi S, Chiriatti A, Petroni S, Schittulli F, Paradiso A. Genetic heterogeneity by comparative genomic hybridization in BRCAx breast cancers. ACTA ACUST UNITED AC 2008; 182:75-83. [DOI: 10.1016/j.cancergencyto.2008.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Revised: 12/21/2007] [Accepted: 01/03/2008] [Indexed: 12/15/2022]
|
23
|
Oldenburg RA, Meijers-Heijboer H, Cornelisse CJ, Devilee P. Genetic susceptibility for breast cancer: How many more genes to be found? Crit Rev Oncol Hematol 2007; 63:125-49. [PMID: 17498966 DOI: 10.1016/j.critrevonc.2006.12.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Revised: 12/01/2006] [Accepted: 12/14/2006] [Indexed: 12/16/2022] Open
Abstract
Today, breast cancer is the most commonly occurring cancer among women. It accounts for 22% of all female cancers and the estimated annual incidence of breast cancer worldwide is about one million cases. Many risk factors have been identified but a positive family history remains among the most important ones established for breast cancer, with first-degree relatives of patients having an approximately two-fold elevated risk. It is currently estimated that approximately 20-25% of this risk is explained by known breast cancer susceptibility genes, mostly those conferring high risks, such as BRCA1 and BRCA2. However, these genes explain less than 5% of the total breast cancer incidence, even though several studies have suggested that the proportion of breast cancer that can be attributed to a genetic factor may be as high as 30%. It is thus likely that there are still breast cancer susceptibility genes to be found. It is presently not known how many such genes there still are, nor how many will fall into the class of rare high-risk (e.g. BRCAx) or of common low-risk susceptibility genes, nor if and how these factors interact with each other to cause susceptibility (a polygenic model). In this review we will address this question and discuss the different undertaken approaches used in identifying new breast cancer susceptibility genes, such as (genome-wide) linkage analysis, CGH, LOH, association studies and global gene expression analysis.
Collapse
Affiliation(s)
- R A Oldenburg
- Center for Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands. r.oldenburg.@erasmusmc.nl
| | | | | | | |
Collapse
|
24
|
Oldenburg RA, Kroeze-Jansema K, Meijers-Heijboer H, van Asperen CJ, Hoogerbrugge N, van Leeuwen I, Vasen HFA, Cleton-Jansen AM, Kraan J, Houwing-Duistermaat JJ, Morreau H, Cornelisse CJ, Devilee P. Characterization of familial non-BRCA1/2 breast tumors by loss of heterozygosity and immunophenotyping. Clin Cancer Res 2006; 12:1693-700. [PMID: 16551851 DOI: 10.1158/1078-0432.ccr-05-2230] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Since the identification of BRCA1 and BRCA2, there has been no major breast cancer susceptibility gene discovered by linkage analysis in breast cancer families. This has been attributed to the heterogeneous genetic basis for the families under study. Recent studies have indicated that breast tumors arising in women carrying a BRCA1 mutation have distinct histopathologic, immunophenotypic, and genetic features. To a lesser extent, this is also true for breast tumors from BRCA2 carriers. This indicates that it might be possible to decrease the genetic heterogeneity among families in which BRCA1 and BRCA2 have been excluded with high certainty (BRCAx families) if distinct subgroups of BRCAx-related breast tumors could be identified. EXPERIMENTAL DESIGN Loss of heterozygosity (LOH) analysis with at least one marker per chromosomal arm (65 markers) was used to characterize 100 breast tumors derived from 92 patients from 42 selected BRCAx families. In addition, the immunophenotype of 10 markers was compared with that of 31 BRCA1- and 21 BRCA2-related breast tumors. RESULTS AND CONCLUSIONS The BRCAx-related tumors were characterized by more frequent LOH at 22q relative to sporadic breast cancer (P < 0.02), and differed significantly from BRCA1- and BRCA2-related tumors in their positivity for Bcl2. However, cluster analyses of the combined data (LOH and immunohistochemistry) did not result in subgroups that would allow meaningful subclassification of the families. On chromosomes 2, 3, 6, 12, 13, 21, and 22, we found markers at which LOH occurred significantly more frequent among the tumors from patients belonging to a single family than expected on the basis of overall LOH frequencies. Nonetheless, linkage analysis with markers for the corresponding regions on chromosomes 12, 21, and 22 did not reveal significant logarithm of the odds.
Collapse
Affiliation(s)
- Rogier A Oldenburg
- Center for Human and Clinical Genetics, Department of Pathology, Leiden University Medical Center, Rotterdam, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Sun X, Zhou Y, Otto KB, Wang M, Chen C, Zhou W, Subramanian K, Vertino PM, Dong JT. Infrequent mutation of ATBF1 in human breast cancer. J Cancer Res Clin Oncol 2006; 133:103-5. [PMID: 16932943 DOI: 10.1007/s00432-006-0148-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2006] [Revised: 07/20/2006] [Accepted: 07/28/2006] [Indexed: 10/24/2022]
Abstract
Deletion at chromosome 16q is frequent in prostate and breast cancers, suggesting the existence of one or more tumor suppressor genes in 16q. Recently, the transcription factor ATBF1 at 16q22 was identified as a strong candidate tumor suppressor gene in prostate cancer, and loss of ATBF1 expression was associated with poorer prognosis in breast cancer. In the present study, we examined mutation, expression, and promoter methylation of ATBF1 in 32 breast cancer cell lines. Only 2 of the 32 cancer cell lines had mutations, although 18 nucleotide polymorphisms were detected. In addition, 24 of 32 (75%) cancer cell lines had reduced ATBF1 mRNA levels, yet promoter methylation was not involved in gene silencing. These findings suggest that ATBF1 plays a role in breast cancer through transcriptional downregulation rather than mutations.
Collapse
Affiliation(s)
- Xiaodong Sun
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Smith P, McGuffog L, Easton DF. A genome wide linkage search for breast cancer susceptibility genes. Genes Chromosomes Cancer 2006; 45:646-55. [PMID: 16575876 PMCID: PMC2714969 DOI: 10.1002/gcc.20330] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Mutations in known breast cancer susceptibility genes account for a minority of the familial aggregation of the disease. To search for further breast cancer susceptibility genes, we performed a combined analysis of four genome-wide linkage screens, which included a total of 149 multiple case breast cancer families. All families included at least three cases of breast cancer diagnosed below age 60 years, at least one of whom had been tested and found not to carry a BRCA1 or BRCA2 mutation. Evidence for linkage was assessed using parametric linkage analysis, assuming both a dominant and a recessive mode of inheritance, and using nonparametric methods. The highest LOD score obtained in any analysis of the combined data was 1.80 under the dominant model, in a region on chromosome 4 close to marker D4S392. Three further LOD scores over 1 were identified in the parametric analyses and two in the nonparametric analyses. A maximum LOD score of 2.40 was found on chromosome arm 2p in families with four or more cases of breast cancer diagnosed below age 50 years. The number of linkage peaks did not differ from the number expected by chance. These results suggest regions that may harbor novel breast cancer susceptibility genes. They also indicate that no single gene is likely to account for a large fraction of the familial aggregation of breast cancer that is not due to mutations in BRCA1 or BRCA2.
Collapse
Affiliation(s)
- Paula Smith
- CR-UKGenetic Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Lesley McGuffog
- CR-UKGenetic Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Douglas F. Easton
- CR-UKGenetic Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| |
Collapse
|
27
|
Abstract
The introduction of comparative genomic hybridization (CGH) in 1992 opened new avenues in genomic investigation; in particular, it advanced analysis of solid tumours, including breast cancer, because it obviated the need to culture cells before their chromosomes could be analyzed. The current generation of CGH analysis uses ordered arrays of genomic DNA sequences and is therefore referred to as array-CGH or matrix-CGH. It was introduced in 1998, and further increased the potential of CGH to provide insight into the fundamental processes of chromosomal instability and cancer. This review provides a critical evaluation of the data published on array-CGH and breast cancer, and discusses some of its expected future value and developments.
Collapse
Affiliation(s)
- Erik H van Beers
- Division of Experimental Therapy, Netherlands Cancer Institute NKI-AVL, Amsterdam
| | - Petra M Nederlof
- Department of Pathology, Netherlands Cancer Institute NKI-AVL, Amsterdam, The Netherlands
| |
Collapse
|