1
|
Espín R, Medina-Jover F, Sigüenza-Andrade J, Farran-Matas S, Mateo F, Figueras A, Sanz R, Vicent G, Shabbir A, Ruiz-Auladell L, Racionero-Andrés E, García I, Baiges A, Franco-Luzón L, Martínez-Tebar A, Pardo-Cea M, Martínez-Iniesta M, Wang X, Cuyàs E, Menendez J, Lopez-Cerda M, Muñoz P, Richaud I, Raya A, Fabregat I, Villanueva A, Serrat X, Cerón J, Alemany M, Guix I, Herencia-Ropero A, Serra V, Krishnan R, Mekhail K, Hakem R, Bruna J, Barcellos-Hoff M, Viñals F, Aytes Á, Pujana M. Harnessing transcriptional regulation of alternative end-joining to predict cancer treatment. NAR Cancer 2025; 7:zcaf007. [PMID: 40061566 PMCID: PMC11886861 DOI: 10.1093/narcan/zcaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/13/2025] [Accepted: 03/04/2025] [Indexed: 04/15/2025] Open
Abstract
Alternative end-joining (alt-EJ) is an error-prone DNA repair pathway that cancer cells deficient in homologous recombination rely on, making them vulnerable to synthetic lethality via inhibition of poly(ADP-ribose) polymerase (PARP). Targeting alt-EJ effector DNA polymerase theta (POLθ), which synergizes with PARP inhibitors and can overcome resistance, is of significant preclinical and clinical interest. However, the transcriptional regulation of alt-EJ and its interactions with processes driving cancer progression remain poorly understood. Here, we show that alt-EJ is suppressed by hypoxia while positively associated with MYC (myelocytomatosis oncogene) transcriptional activity. Hypoxia reduces PARP1 and POLQ expression, decreases MYC binding at their promoters, and lowers PARylation and alt-EJ-mediated DNA repair in cancer cells. Tumors with HIF1A mutations overexpress the alt-EJ gene signature. Inhibition of hypoxia-inducible factor 1α or HIF1A expression depletion, combined with PARP or POLθ inhibition, synergistically reduces the colony-forming capacity of cancer cells. Deep learning reveals the anticorrelation between alt-EJ and hypoxia across regions in tumor images, and the predictions for these and MYC activity achieve area under the curve values between 0.70 and 0.86. These findings further highlight the critical role of hypoxia in modulating DNA repair and present a strategy for predicting and improving outcomes centered on targeting alt-EJ.
Collapse
Affiliation(s)
- Roderic Espín
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Ferran Medina-Jover
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Department of Physiological Sciences, University of Barcelona, L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Javier Sigüenza-Andrade
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Sònia Farran-Matas
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Francesca Mateo
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Agnes Figueras
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Rosario T Sanz
- Molecular Biology Institute of Barcelona, Spanish National Research Council (IBMB-CSIC), Barcelona 08028, Spain
| | - Guillermo Pablo Vicent
- Molecular Biology Institute of Barcelona, Spanish National Research Council (IBMB-CSIC), Barcelona 08028, Spain
| | - Arzoo Shabbir
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Lara Ruiz-Auladell
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | | | - Irene García
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Alexandra Baiges
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Lídia Franco-Luzón
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Adrián Martínez-Tebar
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Miguel Angel Pardo-Cea
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - María Martínez-Iniesta
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Xieng Chen Wang
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Elisabet Cuyàs
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Girona Biomedical Research Institute (IDIBGI), Salt, Girona 17190, Spain
| | - Javier A Menendez
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Girona Biomedical Research Institute (IDIBGI), Salt, Girona 17190, Spain
| | - Marta Lopez-Cerda
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Purificacion Muñoz
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Ivonne Richaud
- Regenerative Medicine Program and Program for Clinical Translation of Regenerative Medicine in Catalonia—P-CMR[C], Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Biomedical Research Network Centre in Bioengineering, Nanomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Angel Raya
- Regenerative Medicine Program and Program for Clinical Translation of Regenerative Medicine in Catalonia—P-CMR[C], Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Biomedical Research Network Centre in Bioengineering, Nanomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona 08010, Spain
| | - Isabel Fabregat
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Biomedical Research Networking Centre in Hepatic and Digestive Diseases (CIBERehd), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Alberto Villanueva
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Xènia Serrat
- Modeling Human Diseases in C. elegans Group, Genes, Diseases, and Therapies Program, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Julián Cerón
- Modeling Human Diseases in C. elegans Group, Genes, Diseases, and Therapies Program, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Montserrat Alemany
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Neuro-Oncology Unit, University Hospital of Bellvitge, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Inés Guix
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Centre, University of California San Francisco, San Francisco, CA 94115, United States
| | - Andrea Herencia-Ropero
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona 08193, Spain
- Experimental Therapeutics Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona 08035, Spain
| | - Violeta Serra
- Experimental Therapeutics Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona 08035, Spain
| | - Rehna Krishnan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Karim Mekhail
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Razqallah Hakem
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jordi Bruna
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Neuro-Oncology Unit, University Hospital of Bellvitge, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Mary Helen Barcellos-Hoff
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Centre, University of California San Francisco, San Francisco, CA 94115, United States
| | - Francesc Viñals
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Department of Physiological Sciences, University of Barcelona, L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Álvaro Aytes
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Miquel Angel Pujana
- ProCURE, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
- Girona Biomedical Research Institute (IDIBGI), Salt, Girona 17190, Spain
| |
Collapse
|
2
|
Jiang W, Wang G, Bai F, Hu B, Xu Y, Xu X, Nie G, Zhu WG, Chen F, Pei XH. BRCA1 Promotes Repair of DNA Damage in Cochlear Hair Cells and Prevents Hearing Loss. J Neurosci 2024; 44:e0132242024. [PMID: 39227158 PMCID: PMC11484548 DOI: 10.1523/jneurosci.0132-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/05/2024] Open
Abstract
Cochlear hair cells (HCs) sense sound waves and allow us to hear. Loss of HCs will cause irreversible sensorineural hearing loss. It is well known that DNA damage repair plays a critical role in protecting cells in many organs. However, how HCs respond to DNA damage and how defective DNA damage repair contributes to hearing loss remain elusive. In this study, we showed that cisplatin induced DNA damage in outer hair cells (OHCs) and promoted OHC loss, leading to hearing loss in mice of either sex. Cisplatin induced the expression of Brca1, a DNA damage repair factor, in OHCs. Deficiency of Brca1 induced OHC and hearing loss, and further promoted cisplatin-induced DNA damage in OHCs, accelerating OHC loss. This study provides the first in vivo evidence demonstrating that cisplatin mainly induces DNA damage in OHCs and that BRCA1 promotes repair of DNA damage in OHCs and prevents hearing loss. Our findings not only demonstrate that DNA damage-inducing agent generates DNA damage in postmitotic HCs but also suggest that DNA repair factors, like BRCA1, protect postmitotic HCs from DNA damage-induced cell death and hearing loss.
Collapse
Affiliation(s)
- Weitao Jiang
- International Cancer Center, Marshall Laboratory of Biomedical Engineering, Department of Otolaryngology, The First Affiliated Hospital, Department of Anatomy and Histology, Shenzhen University Medical School, Shenzhen 518060, China
| | - Guanrun Wang
- International Cancer Center, Marshall Laboratory of Biomedical Engineering, Department of Otolaryngology, The First Affiliated Hospital, Department of Anatomy and Histology, Shenzhen University Medical School, Shenzhen 518060, China
| | - Feng Bai
- Department of Pathology, Shenzhen University Medical School, Shenzhen 518060, China
| | - Bing Hu
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Yang Xu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xingzhi Xu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and International Cancer Center, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Guohui Nie
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University Medical School, Shenzhen 518060, China
| | - Fangyi Chen
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xin-Hai Pei
- International Cancer Center, Marshall Laboratory of Biomedical Engineering, Department of Otolaryngology, The First Affiliated Hospital, Department of Anatomy and Histology, Shenzhen University Medical School, Shenzhen 518060, China
| |
Collapse
|
3
|
Qian T, Bai F, Zhang S, Xu Y, Wang Y, Yuan S, Liu X, Du Y, Peng B, Zhu WG, Xu X, Pei XH. USP11 deubiquitinates E-cadherin and maintains the luminal fate of mammary tumor cells to suppress breast cancer. J Biol Chem 2024; 300:107768. [PMID: 39270819 PMCID: PMC11497446 DOI: 10.1016/j.jbc.2024.107768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 08/01/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Basal-like breast cancer may originate from luminal epithelial or cancerous cells. Inadequately repaired DNA damage impairs luminal differentiation and promotes aberrant luminal to basal trans-differentiation in mammary epithelial cells (MECs). Ubiquitin-specific peptidase 11 (USP11), a deubiquitinase, plays a critical role in DNA damage repair. The role of USP11 in controlling mammary cell differentiation and tumorigenesis remains poorly understood. We generated Usp11 knockout mice and breast cancer cell lines expressing wild-type (WT) and mutant forms of USP11. By using these mutant mice, cell lines, and human USP11-deficient and -proficient breast cancer tissues, we tested how USP11 controls mammary cell fate. We generated Usp11 knock-out mice and found that deletion of Usp11 reduced the expression of E-cadherin and promoted DNA damage in MECs. Overexpression of WT USP11, but not a deubiquitinase-inactive mutant form of USP11, promoted luminal differentiation, enhanced DNA damage repair, and suppressed tumorigenesis in mice. Mechanistically, we found that USP11 enhanced the protein expression of E-cadherin dependent on its deubiquitinase activity and that USP11 deubiquitinated E-cadherin at K738. We discovered that USP11 is bound to E-cadherin through its C-terminal region. In human breast cancers, expression of USP11 was positively correlated with that of E-cadherin, and high USP11 predicted better recurrence-free survival. Our findings provide compelling genetic and biochemical evidence that USP11 not only promotes DNA damage repair but also deubiquitinates E-cadherin and maintains the luminal feature of mammary tumor cells, thereby suppressing luminal breast cancer.
Collapse
Affiliation(s)
- Tao Qian
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Medical School, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
| | - Feng Bai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Medical School, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China; Department of Pathology, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Shiwen Zhang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Medical School, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
| | - Yuping Xu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Medical School, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
| | - Yuchan Wang
- Gansu Dian Medical Laboratory, Lanzhou, China
| | - Shuping Yuan
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Xiong Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Medical School, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
| | - Yaru Du
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Medical School, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
| | - Bin Peng
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Xingzhi Xu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China.
| | - Xin-Hai Pei
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Medical School, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China; Department of Anatomy and Histology, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China.
| |
Collapse
|
4
|
Faraldo MM, Romagnoli M, Wallon L, Dubus P, Deugnier MA, Fre S. Alpha-6 integrin deletion delays the formation of Brca1/p53-deficient basal-like breast tumors by restricting luminal progenitor cell expansion. Breast Cancer Res 2024; 26:91. [PMID: 38835038 PMCID: PMC11151721 DOI: 10.1186/s13058-024-01851-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 05/28/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND The aberrant amplification of mammary luminal progenitors is at the origin of basal-like breast cancers associated with BRCA1 mutations. Integrins mediate cell-matrix adhesion and transmit mechanical and chemical signals that drive epithelial stem cell functions and regulate tumor progression, metastatic reactivation, and resistance to targeted therapies. Consistently, we have recently shown that laminin-binding integrins are essential for the expansion and differentiation of mammary luminal progenitors in physiological conditions. As over-expression of the laminin-binding α6 integrin (Itgα6) is associated with poor prognosis and reduced survival in breast cancer, we here investigate the role of Itgα6 in mammary tumorigenesis. METHODS We used Blg-Cre; Brca1F/F; Trp53F/F mice, a model that phenocopies human basal-like breast cancer with BRCA1 mutations. We generated mutant mice proficient or deficient in Itgα6 expression and followed tumor formation. Mammary tumors and pretumoral tissues were characterized by immunohistochemistry, flow cytometry, RT-qPCR, Western blotting and organoid cultures. Clonogenicity of luminal progenitors from preneoplastic glands was studied in 3D Matrigel cultures. RESULTS We show that Itga6 deletion favors activation of p16 cell cycle inhibitor in the preneoplastic tissue. Subsequently, the amplification of luminal progenitors, the cell of origin of Brca1-deficient tumors, is restrained in Itgα6-deficient gland. In addition, the partial EMT program operating in Brca1/p53-deficient epithelium is attenuated in the absence of Itgα6. As a consequence of these events, mammary tumor formation is delayed in Itgα6-deficient mice. After tumor formation, the lack of Itgα6 does not affect tumor growth but rather alters their differentiation, resulting in reduced expression of basal cell markers. CONCLUSIONS Our data indicate that Itgα6 has a pro-tumorigenic role in Blg-Cre; Brca1F/F; Trp53F/F mice developing basal-like mammary tumors. In particular, we reveal that Itgα6 is required for the luminal progenitor expansion and the aberrant partial EMT program that precedes the formation of BRCA1 deficient tumors.
Collapse
Affiliation(s)
- Marisa M Faraldo
- Laboratory of Genetics and Developmental Biology, Institut Curie, INSERM U934, CNRS UMR3215, PSL Research University, 75248, Paris, France.
| | - Mathilde Romagnoli
- Laboratory of Cell Biology and Cancer, CNRS UMR144, Institut Curie, PSL Research University, 75248, Paris, France
- Institut de Recherches Internationales Servier, 91190, Gif Sur Yvette, France
| | - Loane Wallon
- Laboratory of Genetics and Developmental Biology, Institut Curie, INSERM U934, CNRS UMR3215, PSL Research University, 75248, Paris, France
- Alacris Theranostics GmbH, 12489, Berlin, Germany
| | - Pierre Dubus
- Department of Histology and Pathology, Centre Hospitalier Universitaire de Bordeaux, 33000, Bordeaux, France
- BRIC U1312, INSERM, Bordeaux Institute of Oncology, Université de Bordeaux, 33000, Bordeaux, France
| | - Marie-Ange Deugnier
- Laboratory of Cell Biology and Cancer, CNRS UMR144, Institut Curie, PSL Research University, 75248, Paris, France
| | - Silvia Fre
- Laboratory of Genetics and Developmental Biology, Institut Curie, INSERM U934, CNRS UMR3215, PSL Research University, 75248, Paris, France.
| |
Collapse
|
5
|
Fei X, Liu J, Xu J, Jing H, Cai Z, Yan J, Wu Z, Li H, Wang Z, Shen Y. Integrating spatial transcriptomics and single-cell RNA-sequencing reveals the alterations in epithelial cells during nodular formation in benign prostatic hyperplasia. J Transl Med 2024; 22:380. [PMID: 38654277 PMCID: PMC11036735 DOI: 10.1186/s12967-024-05212-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 04/16/2024] [Indexed: 04/25/2024] Open
Abstract
OBJECTIVE Proliferative nodular formation represents a characteristic pathological feature of benign prostatic hyperplasia (BPH) and serves as the primary cause for prostate volume enlargement and consequent lower urinary tract symptoms (LUTS). Its specific mechanism is largely unknown, although several cellular processes have been reported to be involved in BPH initiation and development and highlighted the crucial role of epithelial cells in proliferative nodular formation. However, the technological limitations hinder the in vivo investigation of BPH patients. METHODS The robust cell type decomposition (RCTD) method was employed to integrate spatial transcriptomics and single cell RNA sequencing profiles, enabling the elucidation of epithelial cell alterations during nodular formation. Immunofluorescent and immunohistochemical staining was performed for verification. RESULTS The alterations of epithelial cells during the formation of nodules in BPH was observed, and a distinct subgroup of basal epithelial (BE) cells, referred to as BE5, was identified to play a crucial role in driving this progression through the hypoxia-induced epithelial-mesenchymal transition (EMT) signaling pathway. BE5 served as both the initiating cell during nodular formation and the transitional cell during the transformation from luminal epithelial (LE) to BE cells. A distinguishing characteristic of the BE5 cell subgroup in patients with BPH was its heightened hypoxia and upregulated expression of FOS. Histological verification results confirmed a significant association between c-Fos expression and key biological processes such as hypoxia and cell proliferation, as well as the close relationship between hypoxia and EMT in BPH tissues. Furthermore, a strong link between c-Fos expression and the progression of BPH was also been validated. Additionally, notable functional differences were observed in glandular and stromal nodules regarding BE5 cells, with BE5 in glandular nodules exhibiting enhanced capacities for EMT and cell proliferation characterized by club-like cell markers. CONCLUSIONS This study elucidated the comprehensive landscape of epithelial cells during in vivo nodular formation in patients, thereby offering novel insights into the initiation and progression of BPH.
Collapse
Affiliation(s)
- Xiawei Fei
- Department of Urology, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, 201799, People's Republic of China
| | - Jican Liu
- Department of Pathology, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, 201799, People's Republic of China
| | - Junyan Xu
- University of Shanghai for Science and Technology, Shanghai, 200093, People's Republic of China
- Department of Urology and Andrology, Gongli Hospital, the Second Military Medical University, Shanghai, 200135, People's Republic of China
| | - Hongyan Jing
- Department of Pathology, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, 201799, People's Republic of China
| | - Zhonglin Cai
- Department of Urology and Andrology, Gongli Hospital, the Second Military Medical University, Shanghai, 200135, People's Republic of China
| | - Jiasheng Yan
- Department of Urology and Andrology, Gongli Hospital, the Second Military Medical University, Shanghai, 200135, People's Republic of China
| | - Zhenqi Wu
- Department of Urology, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, 201799, People's Republic of China
| | - Huifeng Li
- Department of Urology, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, 201799, People's Republic of China.
| | - Zhong Wang
- Department of Urology and Andrology, Gongli Hospital, the Second Military Medical University, Shanghai, 200135, People's Republic of China.
| | - Yanting Shen
- Department of Urology and Andrology, Gongli Hospital, the Second Military Medical University, Shanghai, 200135, People's Republic of China.
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
6
|
Wang X, Bai F, Liu X, Peng B, Xu X, Zhang H, Fu L, Zhu WG, Wang B, Pei XH. GATA3 functions downstream of BRCA1 to promote DNA damage repair and suppress dedifferentiation in breast cancer. BMC Biol 2024; 22:85. [PMID: 38627785 PMCID: PMC11020915 DOI: 10.1186/s12915-024-01881-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 04/04/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Inadequate DNA damage repair promotes aberrant differentiation of mammary epithelial cells. Mammary luminal cell fate is mainly determined by a few transcription factors including GATA3. We previously reported that GATA3 functions downstream of BRCA1 to suppress aberrant differentiation in breast cancer. How GATA3 impacts DNA damage repair preventing aberrant cell differentiation in breast cancer remains elusive. We previously demonstrated that loss of p18, a cell cycle inhibitor, in mice induces luminal-type mammary tumors, whereas depletion of either Brca1 or Gata3 in p18 null mice leads to basal-like breast cancers (BLBCs) with activation of epithelial-mesenchymal transition (EMT). We took advantage of these mutant mice to examine the role of Gata3 as well as the interaction of Gata3 and Brca1 in DNA damage repair in mammary tumorigenesis. RESULTS Depletion of Gata3, like that of Brca1, promoted DNA damage accumulation in breast cancer cells in vitro and in basal-like breast cancers in vivo. Reconstitution of Gata3 improved DNA damage repair in Brca1-deficient mammary tumorigenesis. Overexpression of GATA3 promoted homologous recombination (HR)-mediated DNA damage repair and restored HR efficiency of BRCA1-deficient cells. Depletion of Gata3 sensitized tumor cells to PARP inhibitor (PARPi), and reconstitution of Gata3 enhanced resistance of Brca1-deficient tumor cells to PARP inhibitor. CONCLUSIONS These results demonstrate that Gata3 functions downstream of BRCA1 to promote DNA damage repair and suppress dedifferentiation in mammary tumorigenesis and progression. Our findings suggest that PARP inhibitors are effective for the treatment of GATA3-deficient BLBCs.
Collapse
Affiliation(s)
- Xuejie Wang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Feng Bai
- Department of Pathology, Shenzhen University Medical School, Shenzhen, 518060, China
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Xiong Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Bin Peng
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and International Cancer Center and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Xingzhi Xu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and International Cancer Center and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Hongquan Zhang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Li Fu
- Department of Pharmacology, Shenzhen University Medical School, Shenzhen, 518039, China
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Bin Wang
- Department of General Surgery, Shenzhen Children's Hospital, Shenzhen, 518038, China.
| | - Xin-Hai Pei
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Medical School, Shenzhen, 518060, China.
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA.
- Department of Anatomy and Histology, Shenzhen University Medical School, Shenzhen, 518060, China.
| |
Collapse
|
7
|
Ren S, Bai F, Schnell V, Stanko C, Ritsch M, Schenk T, Barth E, Marz M, Wang B, Pei XH, Bierhoff H. PAPAS promotes differentiation of mammary epithelial cells and suppresses breast carcinogenesis. Cell Rep 2024; 43:113644. [PMID: 38180837 DOI: 10.1016/j.celrep.2023.113644] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 10/18/2023] [Accepted: 12/19/2023] [Indexed: 01/07/2024] Open
Abstract
Extensive remodeling of the female mammary epithelium during development and pregnancy has been linked to cancer susceptibility. The faithful response of mammary epithelial cells (MECs) to hormone signaling is key to avoiding breast cancer development. Here, we show that lactogenic differentiation of murine MECs requires silencing of genes encoding ribosomal RNA (rRNA) by the antisense transcript PAPAS. Accordingly, knockdown of PAPAS derepresses rRNA genes, attenuates the response to lactogenic hormones, and induces malignant transformation. Restoring PAPAS levels in breast cancer cells reduces tumorigenicity and lung invasion and activates many interferon-regulated genes previously linked to metastasis suppression. Mechanistically, PAPAS transcription depends on R-loop formation at the 3' end of rRNA genes, which is repressed by RNase H1 and replication protein A (RPA) overexpression in breast cancer cells. Depletion of PAPAS and upregulation of RNase H1 and RPA in human breast cancer underpin the clinical relevance of our findings.
Collapse
Affiliation(s)
- Sijia Ren
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Hans-Knöll-Str. 2, 07745 Jena, Germany; Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, Department of Anatomy and Histology, Shenzhen University Medical School, Shenzhen 518060, China; Leibniz-Institute on Aging-Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745 Jena, Germany
| | - Feng Bai
- Department of Pathology, Shenzhen University Medical School, Shenzhen 518060, China
| | - Viviane Schnell
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Hans-Knöll-Str. 2, 07745 Jena, Germany; Leibniz-Institute on Aging-Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745 Jena, Germany
| | - Clara Stanko
- Department of Hematology and Medical Oncology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital, Jena, Hans-Knöll-Str. 2, 07745 Jena, Germany
| | - Muriel Ritsch
- Bioinformatics Core Facility Jena, Friedrich Schiller University Jena, Leutragraben 1, 07743 Jena, Germany; RNA Bioinformatics/High Throughput Analysis, Faculty of Mathematics and Computer Science, Leutragraben 1, 07743 Jena, Germany
| | - Tino Schenk
- Department of Hematology and Medical Oncology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital, Jena, Hans-Knöll-Str. 2, 07745 Jena, Germany
| | - Emanuel Barth
- Bioinformatics Core Facility Jena, Friedrich Schiller University Jena, Leutragraben 1, 07743 Jena, Germany; RNA Bioinformatics/High Throughput Analysis, Faculty of Mathematics and Computer Science, Leutragraben 1, 07743 Jena, Germany
| | - Manja Marz
- Bioinformatics Core Facility Jena, Friedrich Schiller University Jena, Leutragraben 1, 07743 Jena, Germany; RNA Bioinformatics/High Throughput Analysis, Faculty of Mathematics and Computer Science, Leutragraben 1, 07743 Jena, Germany
| | - Bin Wang
- Department of General Surgery, Shenzhen Children's Hospital, Shenzhen 518060, China
| | - Xin-Hai Pei
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, Department of Anatomy and Histology, Shenzhen University Medical School, Shenzhen 518060, China.
| | - Holger Bierhoff
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Hans-Knöll-Str. 2, 07745 Jena, Germany; Leibniz-Institute on Aging-Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745 Jena, Germany.
| |
Collapse
|
8
|
Wicker MN, Wagner KU. Cellular Plasticity in Mammary Gland Development and Breast Cancer. Cancers (Basel) 2023; 15:5605. [PMID: 38067308 PMCID: PMC10705338 DOI: 10.3390/cancers15235605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/17/2023] [Accepted: 11/23/2023] [Indexed: 02/12/2024] Open
Abstract
Cellular plasticity is a phenomenon where cells adopt different identities during development and tissue homeostasis as a response to physiological and pathological conditions. This review provides a general introduction to processes by which cells change their identity as well as the current definition of cellular plasticity in the field of mammary gland biology. Following a synopsis of the evolving model of the hierarchical development of mammary epithelial cell lineages, we discuss changes in cell identity during normal mammary gland development with particular emphasis on the effect of the gestation cycle on the emergence of new cellular states. Next, we summarize known mechanisms that promote the plasticity of epithelial lineages in the normal mammary gland and highlight the importance of the microenvironment and extracellular matrix. A discourse of cellular reprogramming during the early stages of mammary tumorigenesis that follows focuses on the origin of basal-like breast cancers from luminal progenitors and oncogenic signaling networks that orchestrate diverse developmental trajectories of transforming epithelial cells. In addition to the epithelial-to-mesenchymal transition, we highlight events of cellular reprogramming during breast cancer progression in the context of intrinsic molecular subtype switching and the genesis of the claudin-low breast cancer subtype, which represents the far end of the spectrum of epithelial cell plasticity. In the final section, we will discuss recent advances in the design of genetically engineered models to gain insight into the dynamic processes that promote cellular plasticity during mammary gland development and tumorigenesis in vivo.
Collapse
Affiliation(s)
| | - Kay-Uwe Wagner
- Department of Oncology, Wayne State University School of Medicine and Tumor Biology Program, Barbara Ann Karmanos Cancer Institute, 4100 John R, EL01TM, Detroit, MI 48201, USA
| |
Collapse
|
9
|
Bai F, Liu X, Zhang X, Mao Z, Wen H, Ma J, Pei XH. p18INK4C and BRCA1 inhibit follicular cell proliferation and dedifferentiation in thyroid cancer. Cell Cycle 2023; 22:1637-1653. [PMID: 37345432 PMCID: PMC10361144 DOI: 10.1080/15384101.2023.2225938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 04/13/2023] [Accepted: 06/09/2023] [Indexed: 06/23/2023] Open
Abstract
Only 3% of thyroid cancers are medullary thyroid carcinomas (MTCs), the rest are follicular epithelial cell derived non-MTCs (NMTCs). A dysfunctional INK4-CDK4-RB pathway is detected in most of NMTCs. DNA repair defects and genome instability are associated with NMTC dedifferentiation and aggressiveness. Whether inactivation of the INK4-CDK4-RB pathway induces NMTCs and how differentiation of NMTC cells is controlled remain elusive. In this study, we generated p18Ink4c and Brca1 singly and doubly deficient mice as well as p16Ink4a and Brca1 singly and doubly deficient mice. By using these mice and human thyroid carcinoma cell lines, we discovered that loss of p18Ink4c, not p16Ink4a, in mice stimulated follicular cell proliferation and induced NMTCs. Depletion of Brca1 alone or both p16Ink4a and Brca1 did not induce thyroid tumor. Depletion of Brca1 in p18Ink4c null mice results in poorly differentiated and aggressive NMTCs with epithelial-mesenchymal transition (EMT) features and enhanced DNA damage. Knockdown of BRCA1 in thyroid carcinoma cells activated EMT and promoted tumorigenesis whereas overexpression of BRCA1 inhibited EMT. BRCA1 and EMT marker expression were inversely related in human thyroid cancers. Our finding, for the first time, demonstrates that inactivation of INK4-CDK4-RB pathway induces NMTCs and that Brca1 deficiency promotes dedifferentiation of NMTC cells. These results suggest that BRCA1 and p18INK4C collaboratively suppress thyroid tumorigenesis and progression and CDK4 inhibitors will be effective for treatment of INK4-inactivated or cyclin D-overexpressed thyroid carcinomas.
Collapse
Affiliation(s)
- Feng Bai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, the First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, China
- Department of Pathology, Shenzhen University Health Science Center, Shenzhen, China
- Dewitt Daughtry Family Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Xiong Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, the First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, China
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, China
| | - Xu Zhang
- Department of Pathology, School of Basic Medicine, Lanzhou University, Lanzhou, China
| | - Zhuo Mao
- Department of Physiology, Shenzhen University Health Science Center, Shenzhen, China
| | - He Wen
- Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Jinshan Ma
- Dewitt Daughtry Family Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
- Department of Thoracic Surgery, Xinjiang Uigur Autonomous Region People’s Hospital, Xinjiang, China
| | - Xin-Hai Pei
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, the First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, China
- Dewitt Daughtry Family Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
10
|
Liu X, Bai F, Wang Y, Wang C, Chan HL, Zheng C, Fang J, Zhu WG, Pei XH. Loss of function of GATA3 regulates FRA1 and c-FOS to activate EMT and promote mammary tumorigenesis and metastasis. Cell Death Dis 2023; 14:370. [PMID: 37353480 PMCID: PMC10290069 DOI: 10.1038/s41419-023-05888-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 06/08/2023] [Accepted: 06/14/2023] [Indexed: 06/25/2023]
Abstract
Basal-like breast cancers (BLBCs) are among the most aggressive cancers, partly due to their enrichment of cancer stem cells (CSCs). Breast CSCs can be generated from luminal-type cancer cells via epithelial-mesenchymal transition (EMT). GATA3 maintains luminal cell fate, and its expression is lost or reduced in BLBCs. However, deletion of Gata3 in mice or cells results in early lethality or proliferative defects. It is unknown how loss-of-function of GATA3 regulates EMT and CSCs in breast cancer. We report here that haploid loss of Gata3 in mice lacking p18Ink4c, a cell cycle inhibitor, up-regulates Fra1, an AP-1 family protein that promotes mesenchymal traits, and downregulates c-Fos, another AP-1 family protein that maintains epithelial fate, leading to activation of EMT and promotion of mammary tumor initiation and metastasis. Depletion of Gata3 in luminal tumor cells similarly regulates Fra1 and c-Fos in activation of EMT. GATA3 binds to FOSL1 (encoding FRA1) and FOS (encoding c-FOS) loci to repress FOSL1 and activate FOS transcription. Deletion of Fra1 or reconstitution of Gata3, but not reconstitution of c-Fos, in Gata3 deficient tumor cells inhibits EMT, preventing tumorigenesis and/or metastasis. In human breast cancers, GATA3 expression is negatively correlated with FRA1 and positively correlated with c-FOS. Low GATA3 and FOS, but high FOSL1, are characteristics of BLBCs. Together, these data provide the first genetic evidence indicating that loss of function of GATA3 in mammary tumor cells activates FOSL1 to promote mesenchymal traits and CSC function, while concurrently repressing FOS to lose epithelial features. We demonstrate that FRA1 is required for the activation of EMT in GATA3 deficient tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Xiong Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Feng Bai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, 518060, China.
- Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China.
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA.
| | - Yuchan Wang
- Gansu Dian Medical Laboratory, Lanzhou, 730000, China
| | - Chuying Wang
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ho Lam Chan
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Chenglong Zheng
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Jian Fang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Xin-Hai Pei
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, 518060, China.
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA.
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, 518060, China.
| |
Collapse
|
11
|
Aylon Y, Furth N, Mallel G, Friedlander G, Nataraj NB, Dong M, Hassin O, Zoabi R, Cohen B, Drendel V, Salame TM, Mukherjee S, Harpaz N, Johnson R, Aulitzky WE, Yarden Y, Shema E, Oren M. Breast cancer plasticity is restricted by a LATS1-NCOR1 repressive axis. Nat Commun 2022; 13:7199. [PMID: 36443319 PMCID: PMC9705295 DOI: 10.1038/s41467-022-34863-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 11/10/2022] [Indexed: 11/29/2022] Open
Abstract
Breast cancer, the most frequent cancer in women, is generally classified into several distinct histological and molecular subtypes. However, single-cell technologies have revealed remarkable cellular and functional heterogeneity across subtypes and even within individual breast tumors. Much of this heterogeneity is attributable to dynamic alterations in the epigenetic landscape of the cancer cells, which promote phenotypic plasticity. Such plasticity, including transition from luminal to basal-like cell identity, can promote disease aggressiveness. We now report that the tumor suppressor LATS1, whose expression is often downregulated in human breast cancer, helps maintain luminal breast cancer cell identity by reducing the chromatin accessibility of genes that are characteristic of a "basal-like" state, preventing their spurious activation. This is achieved via interaction of LATS1 with the NCOR1 nuclear corepressor and recruitment of HDAC1, driving histone H3K27 deacetylation near NCOR1-repressed "basal-like" genes. Consequently, decreased expression of LATS1 elevates the expression of such genes and facilitates slippage towards a more basal-like phenotypic identity. We propose that by enforcing rigorous silencing of repressed genes, the LATS1-NCOR1 axis maintains luminal cell identity and restricts breast cancer progression.
Collapse
Affiliation(s)
- Yael Aylon
- grid.13992.300000 0004 0604 7563Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Noa Furth
- grid.13992.300000 0004 0604 7563Department of Immunology and Regenerative Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Giuseppe Mallel
- grid.13992.300000 0004 0604 7563Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Gilgi Friedlander
- grid.13992.300000 0004 0604 7563Department of Life Sciences Core Facilities, The Nancy & Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Nishanth Belugali Nataraj
- grid.13992.300000 0004 0604 7563Department of Immunology and Regenerative Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Meng Dong
- grid.502798.10000 0004 0561 903XDr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology and University of Tuebingen, Stuttgart, Germany
| | - Ori Hassin
- grid.13992.300000 0004 0604 7563Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Rawan Zoabi
- grid.13992.300000 0004 0604 7563Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Benjamin Cohen
- grid.13992.300000 0004 0604 7563Department of Immunology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Vanessa Drendel
- grid.416008.b0000 0004 0603 4965Department of Pathology, Robert Bosch Hospital, Stuttgart, Germany
| | - Tomer Meir Salame
- grid.13992.300000 0004 0604 7563Flow Cytometry Unit, Department of Life Sciences Core Facilities, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Saptaparna Mukherjee
- grid.13992.300000 0004 0604 7563Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Nofar Harpaz
- grid.13992.300000 0004 0604 7563Department of Immunology and Regenerative Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Randy Johnson
- grid.240145.60000 0001 2291 4776Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Walter E. Aulitzky
- grid.416008.b0000 0004 0603 4965Department of Hematology, Oncology and Palliative Medicine, Robert Bosch Hospital, Stuttgart, Germany
| | - Yosef Yarden
- grid.13992.300000 0004 0604 7563Department of Immunology and Regenerative Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Efrat Shema
- grid.13992.300000 0004 0604 7563Department of Immunology and Regenerative Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Moshe Oren
- grid.13992.300000 0004 0604 7563Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| |
Collapse
|
12
|
Loss of function of BRCA1 promotes EMT in mammary tumors through activation of TGFβR2 signaling pathway. Cell Death Dis 2022; 13:195. [PMID: 35236825 PMCID: PMC8891277 DOI: 10.1038/s41419-022-04646-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 01/25/2022] [Accepted: 02/09/2022] [Indexed: 12/15/2022]
Abstract
BRCA1 deficient breast cancers are aggressive and chemoresistant due, in part, to their enrichment of cancer stem cells that can be generated from carcinoma cells by an epithelial-mesenchymal transition (EMT). We previously discovered that BRCA1 deficiency activates EMT in mammary tumorigenesis. How BRCA1 controls EMT and how to effectively target BRCA1-deficient cancers remain elusive. We analyzed murine and human tumors and identified a role for Tgfβr2 in governing the molecular aspects of EMT that occur with Brca1 loss. We utilized CRISPR to delete Tgfβr2 and specific inhibitors to block Tgfβr2 activity and followed up with the molecular analysis of assays for tumor growth and metastasis. We discovered that heterozygous germline deletion, or epithelia-specific deletion of Brca1 in mice, activates Tgfβr2 signaling pathways in mammary tumors. BRCA1 depletion promotes TGFβ-mediated EMT activation in cancer cells. BRCA1 binds to the TGFβR2 locus to repress its transcription. Targeted deletion or pharmaceutical inhibition of Tgfβr2 in Brca1-deficient tumor cells reduces EMT and suppresses tumorigenesis and metastasis. BRCA1 and TGFβR2 expression levels are inversely related in human breast cancers. This study reveals for the first time that a targetable TGFβR signaling pathway is directly activated by BRCA1-deficiency in the induction of EMT in breast cancer progression.
Collapse
|
13
|
Abstract
Triple-negative breast cancer (TNBC) encompasses a heterogeneous group of fundamentally different diseases with different histologic, genomic, and immunologic profiles, which are aggregated under this term because of their lack of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 expression. Massively parallel sequencing and other omics technologies have demonstrated the level of heterogeneity in TNBCs and shed light into the pathogenesis of this therapeutically challenging entity in breast cancer. In this review, we discuss the histologic and molecular classifications of TNBC, the genomic alterations these different tumor types harbor, and the potential impact of these alterations on the pathogenesis of these tumors. We also explore the role of the tumor microenvironment in the biology of TNBCs and its potential impact on therapeutic response. Dissecting the biology and understanding the therapeutic dependencies of each TNBC subtype will be essential to delivering on the promise of precision medicine for patients with triple-negative disease.
Collapse
Affiliation(s)
- Fatemeh Derakhshan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA;
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA;
| |
Collapse
|
14
|
Bai F, Zheng C, Liu X, Chan HL, Liu S, Ma J, Ren S, Zhu WG, Pei XH. Loss of function of GATA3 induces basal-like mammary tumors. Am J Cancer Res 2022; 12:720-733. [PMID: 34976209 PMCID: PMC8692904 DOI: 10.7150/thno.65796] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/13/2021] [Indexed: 11/25/2022] Open
Abstract
Purpose: GATA3 is a transcription factor essential for mammary luminal epithelial cell differentiation. Expression of GATA3 is absent or significantly reduced in basal-like breast cancers. Gata3 loss-of-function impairs cell proliferation, making it difficult to investigate the role of GATA3 deficiency in vivo. We previously demonstrated that CDK inhibitor p18INK4c (p18) is a downstream target of GATA3 and restrains mammary epithelial cell proliferation and tumorigenesis. Whether and how loss-of-function of GATA3 results in basal-like breast cancers remains elusive. Methods: We generated mutant mouse strains with heterozygous germline deletion of Gata3 in p18 deficient backgrounds and developed a Gata3 depleted mammary tumor model system to determine the role of Gata3 loss in controlling cell proliferation and aberrant differentiation in mammary tumor development and progression. Results: Haploid loss of Gata3 reduced mammary epithelial cell proliferation with induction of p18, impaired luminal differentiation, and promoted basal differentiation in mammary glands. p18 deficiency induced luminal type mammary tumors and rescued the proliferative defect caused by haploid loss of Gata3. Haploid loss of Gata3 accelerated p18 deficient mammary tumor development and changed the properties of these tumors, resulting in their malignant and luminal-to-basal transformation. Expression of Gata3 negatively correlated with basal differentiation markers in MMTV-PyMT mammary tumor cells. Depletion of Gata3 in luminal tumor cells also reduced cell proliferation with induction of p18 and promoted basal differentiation. We confirmed that expression of GATA3 and basal markers are inversely correlated in human basal-like breast cancers. Conclusions: This study provides the first genetic evidence demonstrating that loss-of-function of GATA3 directly induces basal-like breast cancer. Our finding suggests that basal-like breast cancer may also originate from luminal type cancer.
Collapse
|
15
|
Bai F, Zhang LH, Liu X, Wang C, Zheng C, Sun J, Li M, Zhu WG, Pei XH. GATA3 functions downstream of BRCA1 to suppress EMT in breast cancer. Theranostics 2021; 11:8218-8233. [PMID: 34373738 PMCID: PMC8344017 DOI: 10.7150/thno.59280] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/05/2021] [Indexed: 02/06/2023] Open
Abstract
Purpose: Functional loss of BRCA1 is associated with poorly differentiated and metastatic breast cancers that are enriched with cancer stem cells (CSCs). CSCs can be generated from carcinoma cells through an epithelial-mesenchymal transition (EMT) program. We and others have previously demonstrated that BRCA1 suppresses EMT and regulates the expression of multiple EMT-related transcription factors. However, the downstream mediators of BRCA1 function in EMT suppression remain elusive. Methods: Depletion of BRCA1 or GATA3 activates p18INK4C , a cell cycle inhibitor which inhibits mammary epithelial cell proliferation. We have therefore created genetically engineered mice with Brca1 or Gata3 loss in addition to deletion of p18INK4C , to rescue proliferative defects caused by deficiency of Brca1 or Gata3. By using these mutant mice along with human BRCA1 deficient as well as proficient breast cancer tissues and cells, we investigated and compared the role of Brca1 and Gata3 loss in the activation of EMT in breast cancers. Results: We discovered that BRCA1 and GATA3 expressions were positively correlated in human breast cancer. Depletion of BRCA1 stimulated methylation of GATA3 promoter thereby repressing GATA3 transcription. We developed Brca1 and Gata3 deficient mouse system. We found that Gata3 deficiency in mice induced poorly-differentiated mammary tumors with the activation of EMT and promoted tumor initiating and metastatic potential. Gata3 deficient mammary tumors phenocopied Brca1 deficient tumors in the induction of EMT under the same genetic background. Reconstitution of Gata3 in Brca1-deficient tumor cells activated mesenchymal-epithelial transition, suppressing tumor initiation and metastasis. Conclusions: Our finding, for the first time, demonstrates that GATA3 functions downstream of BRCA1 to suppress EMT in controlling mammary tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Feng Bai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
- Department of Pathology, Shenzhen University Health Science Center, Shenzhen 518060, China
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136, USA
| | - Li-Han Zhang
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136, USA
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000, China
- The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, China
| | - Xiong Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Chuying Wang
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136, USA
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Chenglong Zheng
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Jianping Sun
- Department of Mathematics and Statistics, University of North Carolina at Greensboro, Greensboro, NC 27402, USA
| | - Min Li
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Xin-Hai Pei
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136, USA
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen 518060, China
| |
Collapse
|
16
|
Hu L, Su L, Cheng H, Mo C, Ouyang T, Li J, Wang T, Fan Z, Fan T, Lin B, Zhang J, Xie Y. Single-Cell RNA Sequencing Reveals the Cellular Origin and Evolution of Breast Cancer in BRCA1 Mutation Carriers. Cancer Res 2021; 81:2600-2611. [PMID: 33727227 DOI: 10.1158/0008-5472.can-20-2123] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/29/2020] [Accepted: 03/11/2021] [Indexed: 11/16/2022]
Abstract
The cell of origin and the development of breast cancer are not fully elucidated in BRCA1 mutation carriers, especially for estrogen receptor (ER)-positive breast cancers. Here, we performed single-cell RNA sequencing (RNA-seq) on 82,122 cells isolated from the breast cancer tissues and adjacent or prophylactic normal breast tissues from four BRCA1 mutation carriers and three noncarriers. Whole-exome sequencing was performed on breast tumors from the four BRCA1 mutation carriers; for validation, bulk RNA-seq was performed on adjacent normal breast tissues from eight additional BRCA1 mutation carriers and 14 noncarriers. Correlation analyses suggested that breast cancers in BRCA1 mutation carriers might originate from luminal cells. The aberrant luminal progenitor cells with impaired differentiation were significantly increased in normal breast tissues in BRCA1 mutation carriers compared with noncarriers. These observations were further validated by the bulk RNA-seq data from additional BRCA1 mutation carriers. These data suggest that the cell of origin of basal-like breast tumors (ERneg) in BRCA1 mutation carriers might be luminal progenitor cells. The expression of TP53 and BRCA1 was decreased in luminal progenitor cells from normal breast tissue in BRCA1 mutation carriers, which might trigger the basal/mesenchymal transition of luminal progenitors and might result in basal-like tumor development. Furthermore, ERhigh luminal tumors might originate from mature luminal cells. Our study provides in-depth evidence regarding the cells of origin of different breast cancer subtypes in BRCA1 mutation carriers. SIGNIFICANCE: Single-cell RNA-seq data indicate that basal-like breast cancer (ERneg) might originate from luminal progenitors, and ERhigh luminal breast cancer might originate from mature luminal cells in BRCA1 mutation carriers.
Collapse
Affiliation(s)
- Li Hu
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Liming Su
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Hainan Cheng
- Berry Oncology Co., Ltd. (Berry Genomics Group), Beijing, P.R. China
| | - Chunling Mo
- Berry Oncology Co., Ltd. (Berry Genomics Group), Beijing, P.R. China
| | - Tao Ouyang
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Jinfeng Li
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Tianfeng Wang
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Zhaoqing Fan
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Tie Fan
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Benyao Lin
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Jianguang Zhang
- Berry Oncology Co., Ltd. (Berry Genomics Group), Beijing, P.R. China.
| | - Yuntao Xie
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P.R. China.
| |
Collapse
|
17
|
Miklikova S, Trnkova L, Plava J, Bohac M, Kuniakova M, Cihova M. The Role of BRCA1/2-Mutated Tumor Microenvironment in Breast Cancer. Cancers (Basel) 2021; 13:575. [PMID: 33540843 PMCID: PMC7867315 DOI: 10.3390/cancers13030575] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/23/2021] [Accepted: 01/29/2021] [Indexed: 12/15/2022] Open
Abstract
Taking into account the factors of high incidence rate, prevalence and mortality, breast cancer represents a crucial social and economic burden. Most cases of breast cancer develop as a consequence of somatic mutations accumulating in mammary epithelial cells throughout lifetime and approximately 5-10% can be ascribed to monogenic predispositions. Even though the role of genetic predispositions in breast cancer is well described in the context of genetics, very little is known about the role of the microenvironment carrying the same aberrant cells impaired by the germline mutation in the breast cancer development and progression. Based on the clinical observations, carcinomas carrying mutations in hereditary tumor-suppressor genes involved in maintaining genome integrity such as BRCA1/2 have worse prognosis and aggressive behavior. One of the mechanisms clarifying the aggressive nature of BRCA-associated tumors implies alterations within the surrounding adipose tissue itself. The objective of this review is to look at the role of BRCA1/2 mutations in the context of breast tumor microenvironment and plausible mechanisms by which it contributes to the aggressive behavior of the tumor cells.
Collapse
Affiliation(s)
- Svetlana Miklikova
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, 84505 Bratislava, Slovakia; (S.M.); (L.T.); (J.P.)
| | - Lenka Trnkova
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, 84505 Bratislava, Slovakia; (S.M.); (L.T.); (J.P.)
| | - Jana Plava
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, 84505 Bratislava, Slovakia; (S.M.); (L.T.); (J.P.)
| | - Martin Bohac
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, National Cancer Institute, Klenova 1, 83310 Bratislava, Slovakia;
- Department of Oncosurgery, National Cancer Institute, Klenova 1, 83310 Bratislava, Slovakia
- Regenmed Ltd., Medena 29, 81108 Bratislava, Slovakia
| | - Marcela Kuniakova
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 81108 Bratislava, Slovakia;
| | - Marina Cihova
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, 84505 Bratislava, Slovakia; (S.M.); (L.T.); (J.P.)
| |
Collapse
|
18
|
Bai F, Liu S, Liu X, Hollern DP, Scott A, Wang C, Zhang L, Fan C, Fu L, Perou CM, Zhu WG, Pei XH. PDGFRβ is an essential therapeutic target for BRCA1-deficient mammary tumors. Breast Cancer Res 2021; 23:10. [PMID: 33478572 PMCID: PMC7819225 DOI: 10.1186/s13058-021-01387-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/03/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Basal-like breast cancers (BLBCs) are a leading cause of cancer death due to their capacity to metastasize and lack of effective therapies. More than half of BLBCs have a dysfunctional BRCA1. Although most BRCA1-deficient cancers respond to DNA-damaging agents, resistance and tumor recurrence remain a challenge to survival outcomes for BLBC patients. Additional therapies targeting the pathways aberrantly activated by BRCA1 deficiency are urgently needed. METHODS Most BRCA1-deficient BLBCs carry a dysfunctional INK4-RB pathway. Thus, we created genetically engineered mice with Brca1 loss and deletion of p16INK4A, or separately p18INK4C, to model the deficient INK4-RB signaling in human BLBC. By using these mutant mice and human BRCA1-deficient and proficient breast cancer tissues and cells, we tested if there exists a druggable target in BRCA1-deficient breast cancers. RESULTS Heterozygous germline or epithelium-specific deletion of Brca1 in p18INK4C- or p16INK4A-deficient mice activated Pdgfrβ signaling, induced epithelial-to-mesenchymal transition, and led to BLBCs. Confirming this role, targeted deletion of Pdgfrβ in Brca1-deficient tumor cells promoted cell death, induced mesenchymal-to-epithelial transition, and suppressed tumorigenesis. Importantly, we also found that pharmaceutical inhibition of Pdgfrβ and its downstream target Pkcα suppressed Brca1-deficient tumor initiation and progression and effectively killed BRCA1-deficient cancer cells. CONCLUSIONS Our work offers the first genetic and biochemical evidence that PDGFRβ-PKCα signaling is repressed by BRCA1, which establishes PDGFRβ-PKCα signaling as a therapeutic target for BRCA1-deficient breast cancers.
Collapse
Affiliation(s)
- Feng Bai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, China
- Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Shiqin Liu
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xiong Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, China
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Daniel P Hollern
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alexandria Scott
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Chuying Wang
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lihan Zhang
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
- The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China
| | - Cheng Fan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Li Fu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, China
- Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, 518039, China
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Xin-Hai Pei
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, China.
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA.
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, 518060, China.
| |
Collapse
|
19
|
Singh AK, Yu X. Tissue-Specific Carcinogens as Soil to Seed BRCA1/2-Mutant Hereditary Cancers. Trends Cancer 2020; 6:559-568. [PMID: 32336659 DOI: 10.1016/j.trecan.2020.03.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 02/06/2023]
Abstract
Despite their ubiquitous expression, the inheritance of monoallelic germline mutations in breast cancer susceptibility gene type 1 or 2 (BRCA1/2) poses tissue-specific variations in cancer risks and primarily associate with familial breast and ovarian cancers. The molecular basis of this tissue-specific tumor incidence remains unknown and intriguing to cancer researchers. A plethora of recent reports support the idea that several nongenetic factors present in the tissue microenvironment could induce tumors in the mutant BRCA1/2 background. This Opinion article summarizes the recent advances on tissue-specific carcinogens and their complex crosstalk with the compromised DNA repair machinery of BRCA1/2-mutant cells. Finally, we present our perspective on the therapeutic and chemopreventive interpretations of these developments.
Collapse
Affiliation(s)
- Anup Kumar Singh
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Xiaochun Yu
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA.
| |
Collapse
|
20
|
Hilton HN, Patterson McDonald LJ, Santucci N, van der Bent FR, Silvestri A, Graham JD, Clarke CL. BRCA1 Attenuates Progesterone Effects on Proliferation and NFκB Activation in Normal Human Mammary Epithelial Cells. J Mammary Gland Biol Neoplasia 2019; 24:257-270. [PMID: 31104199 DOI: 10.1007/s10911-019-09431-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 04/21/2019] [Indexed: 12/25/2022] Open
Abstract
Germline mutations in the breast cancer susceptibility gene BRCA1, encoding a tumor suppressor protein, greatly enhance the risk of breast and ovarian cancer. This tissue-specificity implicates the role of ovarian hormones. Indeed, BRCA1 has been demonstrated to regulate the signalling axis of the hormone, progesterone, and its receptor, the progesterone receptor (PR), and progesterone action has been implicated in BRCA1-related tumorigenesis. BRCA1 also plays important roles in oxidative stress and activating nuclear factor kappaB (NFκB) signalling pathways. Like wildtype BRCA1 function, PR signalling has also been shown to inhibit NFκB activation. Although PR and BRCA1 networks are known to interact, their interaction at the level of NFκB activation in the human breast is not understood. This study investigates the effect of reduced BRCA1 expression on proliferation and NFκB activation in human breast cells, and the impact of progesterone on these effects. The major findings are that: 1) Reduced BRCA1 levels inhibit cell growth in normal human mammary cells and breast cancer cells; 2) Reduced BRCA1 levels stimulated inflammatory targets and NFκB activity in normal human mammary cells; 3) Wildtype BRCA1 inhibited the pro-proliferative effects of progesterone in normal mammary epithelial cells, and; 4) Progesterone attenuated BRCA1-mediated NFκB activation in normal human mammary cells. These data have important implications for our understanding of progesterone action in BRCA1 mutation carriers, and how inhibition of this action may potentially delay tumorigenesis or impart a more favourable prognosis.
Collapse
Affiliation(s)
- H N Hilton
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney Medical School - Westmead, The University of Sydney, Westmead, NSW, 2145, Australia
| | - L J Patterson McDonald
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney Medical School - Westmead, The University of Sydney, Westmead, NSW, 2145, Australia
| | - N Santucci
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney Medical School - Westmead, The University of Sydney, Westmead, NSW, 2145, Australia
| | - F R van der Bent
- Department of Medicine, Academic Medical Center, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - A Silvestri
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney Medical School - Westmead, The University of Sydney, Westmead, NSW, 2145, Australia
| | - J D Graham
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney Medical School - Westmead, The University of Sydney, Westmead, NSW, 2145, Australia.
| | - C L Clarke
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney Medical School - Westmead, The University of Sydney, Westmead, NSW, 2145, Australia
| |
Collapse
|
21
|
Zavala VA, Serrano-Gomez SJ, Dutil J, Fejerman L. Genetic Epidemiology of Breast Cancer in Latin America. Genes (Basel) 2019; 10:E153. [PMID: 30781715 PMCID: PMC6410045 DOI: 10.3390/genes10020153] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 12/20/2022] Open
Abstract
The last 10 years witnessed an acceleration of our understanding of what genetic factors underpin the risk of breast cancer. Rare high- and moderate-penetrance variants such as those in the BRCA genes account for a small proportion of the familial risk of breast cancer. Low-penetrance alleles are expected to underlie the remaining heritability. By now, there are about 180 genetic polymorphisms that are associated with risk, most of them of modest effect. In combination, they can be used to identify women at the lowest or highest ends of the risk spectrum, which might lead to more efficient cancer prevention strategies. Most of these variants were discovered in populations of European descent. As a result, we might be failing to discover additional polymorphisms that could explain risk in other groups. This review highlights breast cancer genetic epidemiology studies conducted in Latin America, and summarizes the information that they provide, with special attention to similarities and differences with studies in other populations. It includes studies of common variants, as well as moderate- and high-penetrance variants. In addition, it addresses the gaps that need to be bridged in order to better understand breast cancer genetic risk in Latin America.
Collapse
Affiliation(s)
- Valentina A Zavala
- Department of Medicine, Division of General Internal Medicine, University of California San Francisco, San Francisco, CA 94143-1793, USA.
| | - Silvia J Serrano-Gomez
- Grupo de investigación en biología del cáncer, Instituto Nacional de Cancerología, Bogotá 11001000, Colombia.
| | - Julie Dutil
- Cancer Biology Division, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR 00732, USA.
| | - Laura Fejerman
- Department of Medicine, Division of General Internal Medicine, University of California San Francisco, San Francisco, CA 94143-1793, USA.
| |
Collapse
|
22
|
Cai M, Zhang H, Hou L, Gao W, Song Y, Cui X, Li C, Guan R, Ma J, Wang X, Han Y, Lv Y, Chen F, Wang P, Meng X, Fu S. Inhibiting homologous recombination decreases extrachromosomal amplification but has no effect on intrachromosomal amplification in methotrexate-resistant colon cancer cells. Int J Cancer 2018; 144:1037-1048. [PMID: 30070702 PMCID: PMC6586039 DOI: 10.1002/ijc.31781] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 02/23/2018] [Accepted: 07/24/2018] [Indexed: 01/08/2023]
Abstract
Gene amplification, which involves the two major topographical structures double minutes (DMs) and homegeneously stained region (HSR), is a common mechanism of treatment resistance in cancer and is initiated by DNA double‐strand breaks. NHEJ, one of DSB repair pathways, is involved in gene amplification as we demonstrated previously. However, the involvement of homologous recombination, another DSB repair pathway, in gene amplification remains to be explored. To better understand the association between HR and gene amplification, we detected HR activity in DM‐ and HSR‐containing MTX‐resistant HT‐29 colon cancer cells. In DM‐containing MTX‐resistant cells, we found increased homologous recombination activity compared with that in MTX‐sensitive cells. Therefore, we suppressed HR activity by silencing BRCA1, the key player in the HR pathway. The attenuation of HR activity decreased the numbers of DMs and DM‐form amplified gene copies and increased the exclusion of micronuclei and nuclear buds that contained DM‐form amplification; these changes were accompanied by cell cycle acceleration and increased MTX sensitivity. In contrast, BRCA1 silencing did not influence the number of amplified genes and MTX sensitivity in HSR‐containing MTX‐resistant cells. In conclusion, our results suggest that the HR pathway plays different roles in extrachromosomal and intrachromosomal gene amplification and may be a new target to improve chemotherapeutic outcome by decreasing extrachromosomal amplification in cancer. What's new? Double‐strand DNA breaks (DSBs) initiate gene amplification, a phenomenon associated with therapeutic resistance in cancer that involves two topographical structures, double minutes (DMs) and homogeneously staining regions (HSRs). Whether DSB repair pathways, particularly homologous recombination (HR), also influence gene amplification is unknown. Here, in methotrexate‐resistant colon cancer cells, HR inhibition effectively reduced gene amplification, specifically the DM‐form, by blocking DM formation and promoting DM exclusion via micronuclei. HR inhibition had no influence on the HSR‐form of gene amplification. Loss of gene amplification by HR inhibition, through partial reversal of methotrexate resistance, may contribute to improved chemotherapeutic outcome.
Collapse
Affiliation(s)
- Mengdi Cai
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| | - Huishu Zhang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| | - Liqing Hou
- Department of Genetics, Inner Mongolia Maternal and Child Care Hospital, Hohhot, Inner Mongolia Autonomous Region, China
| | - Wei Gao
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| | - Ying Song
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| | - Xiaobo Cui
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| | - Chunxiang Li
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| | - Rongwei Guan
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| | - Jinfa Ma
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| | - Xu Wang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| | - Yue Han
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| | - Yafan Lv
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| | - Feng Chen
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| | - Ping Wang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| | - Xiangning Meng
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| | - Songbin Fu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, 150081, China
| |
Collapse
|
23
|
Dittmer J. Breast cancer stem cells: Features, key drivers and treatment options. Semin Cancer Biol 2018; 53:59-74. [PMID: 30059727 DOI: 10.1016/j.semcancer.2018.07.007] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/10/2018] [Accepted: 07/18/2018] [Indexed: 02/06/2023]
Abstract
The current view is that breast cancer is a stem cell disease characterized by the existence of cancer cells with stem-like features and tumor-initiating potential. These cells are made responsible for tumor dissemination and metastasis. Common therapies by chemotherapeutic drugs fail to eradicate these cells and rather increase the pool of cancer stem cells in tumors, an effect that may increase the likelyhood of recurrence. Fifteen years after the first evidence for a small stem-like subpopulation playing a major role in breast cancer initiation has been published a large body of knowledge has been accumulated regarding the signaling cascades and proteins involved in maintaining stemness in breast cancer. Differences in the stem cell pool size and in mechanisms regulating stemness in the different breast cancer subtypes have emerged. Overall, this knowledge offers new approaches to intervene with breast cancer stem cell activity. New options are particularly needed for the treatment of triple-negative breast cancer subtype, which is particularly rich in cancer stem cells and is also the subtype for which specific therapies are still not available.
Collapse
Affiliation(s)
- Jürgen Dittmer
- Clinic for Gynecology, Martin Luther University Halle-Wittenberg, Germany.
| |
Collapse
|
24
|
Wang C, Bai F, Zhang LH, Scott A, Li E, Pei XH. Estrogen promotes estrogen receptor negative BRCA1-deficient tumor initiation and progression. Breast Cancer Res 2018; 20:74. [PMID: 29996906 PMCID: PMC6042319 DOI: 10.1186/s13058-018-0996-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/30/2018] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Estrogen promotes breast cancer development and progression mainly through estrogen receptor (ER). However, blockage of estrogen production or action prevents development of and suppresses progression of ER-negative breast cancers. How estrogen promotes ER-negative breast cancer development and progression is poorly understood. We previously discovered that deletion of cell cycle inhibitors p16Ink4a (p16) or p18Ink4c (p18) is required for development of Brca1-deficient basal-like mammary tumors, and that mice lacking p18 develop luminal-type mammary tumors. METHODS A genetic model system with three mouse strains, one that develops ER-positive mammary tumors (p18 single deletion) and the others that develop ER-negative tumors (p16;Brca1 and p18;Brca1 compound deletion), human BRCA1 mutant breast cancer patient-derived xenografts, and human BRCA1-deficient and BRCA1-proficient breast cancer cells were used to determine the role of estrogen in activating epithelial-mesenchymal transition (EMT), stimulating cell proliferation, and promoting ER-negative mammary tumor initiation and metastasis. RESULTS Estrogen stimulated the proliferation and tumor-initiating potential of both ER-positive Brca1-proficient and ER-negative Brca1-deficient tumor cells. Estrogen activated EMT in a subset of Brca1-deficient mammary tumor cells that maintained epithelial features, and enhanced the number of cancer stem cells, promoting tumor progression and metastasis. Estrogen activated EMT independent of ER in Brca1-deficient, but not Brca1-proficient, tumor cells. Estrogen activated the AKT pathway in BRCA1-deficient tumor cells independent of ER, and pharmaceutical inhibition of AKT activity suppressed EMT and cell proliferation preventing BRCA1 deficient tumor progression. CONCLUSIONS This study reveals for the first time that estrogen promotes BRCA1-deficient tumor initiation and progression by stimulation of cell proliferation and activation of EMT, which are dependent on AKT activation and independent of ER.
Collapse
Affiliation(s)
- Chuying Wang
- Department of Medical Oncology, The First Affiliated hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 People’s Republic of China
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136 USA
| | - Feng Bai
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136 USA
| | - Li-han Zhang
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136 USA
| | - Alexandria Scott
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136 USA
| | - Enxiao Li
- Department of Medical Oncology, The First Affiliated hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 People’s Republic of China
| | - Xin-Hai Pei
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136 USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136 USA
| |
Collapse
|
25
|
Hong D, Fritz AJ, Zaidi SK, van Wijnen AJ, Nickerson JA, Imbalzano AN, Lian JB, Stein JL, Stein GS. Epithelial-to-mesenchymal transition and cancer stem cells contribute to breast cancer heterogeneity. J Cell Physiol 2018; 233:9136-9144. [PMID: 29968906 DOI: 10.1002/jcp.26847] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 05/01/2018] [Indexed: 12/17/2022]
Abstract
Breast cancer is the most common cancer in women, and accounts for ~30% of new cancer cases and 15% of cancer-related deaths. Tumor relapse and metastasis are primary factors contributing to breast cancer-related deaths. Therefore, the challenge for breast cancer treatment is to sustain remission. A driving force behind tumor relapse is breast cancer heterogeneity (both intertumor, between different patients, and intratumor, within the same tumor). Understanding breast cancer heterogeneity is necessary to develop preventive interventions and targeted therapies. A recently emerging concept is that intratumor heterogeneity is driven by cancer stem cells (CSCs) that are capable of giving rise to a multitude of different cells within a tumor. Studies have highlighted linkage of CSC formation with epithelial-to-mesenchymal transition (EMT). In this review, we summarize the current understanding of breast cancer heterogeneity, links between EMT and CSCs, regulation of EMT by Runx transcription factors, and potential therapeutic strategies targeting these processes.
Collapse
Affiliation(s)
- Deli Hong
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont, Burlington, Vermont
| | - Andrew J Fritz
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont, Burlington, Vermont
| | - Sayyed K Zaidi
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont, Burlington, Vermont
| | - Andre J van Wijnen
- Departments of Orthopedic Surgery and Biochemistry & Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | | | - Anthony N Imbalzano
- Graduate Program in Cell Biology and Department of Biochemistry and Molecular Pharmacology, UMass Medical School, Worcester, Massachusetts
| | - Jane B Lian
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont, Burlington, Vermont
| | - Janet L Stein
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont, Burlington, Vermont
| | - Gary S Stein
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont, Burlington, Vermont
| |
Collapse
|
26
|
Liu S, Chan HL, Bai F, Ma J, Scott A, Robbins DJ, Capobianco AJ, Zhu P, Pei XH. Gata3 restrains B cell proliferation and cooperates with p18INK4c to repress B cell lymphomagenesis. Oncotarget 2018; 7:64007-64020. [PMID: 27588406 PMCID: PMC5325421 DOI: 10.18632/oncotarget.11746] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 08/24/2016] [Indexed: 12/13/2022] Open
Abstract
GATA3, a lineage specifier, controls lymphoid cell differentiation and its function in T cell commitment and development has been extensively studied. GATA3 promotes T cell specification by repressing B cell potential in pro T cells and decreased GATA3 expression is essential for early B cell commitment. Inherited genetic variation in GATA3 has been associated with lymphoma susceptibility. However, it remains elusive how the loss of function of GATA3 promotes B cell development and induces B cell lymphomas. In this study, we found that haploid loss of Gata3 by heterozygous germline deletion increased B cell populations in the bone marrow (BM) and spleen, and decreased CD4 T cell populations in the thymus, confirming that Gata3 promotes T and suppresses B cell development. We discovered that haploid loss of Gata3 reduced thymocyte proliferation with induction of p18Ink4c (p18), an inhibitor of CDK4 and CDK6, but enhanced B cell proliferation in the BM and spleen independent of p18. Loss of p18 partially restored Gata3 deficient thymocyte proliferation, but further stimulated Gata3 deficient B cell proliferation in the BM and spleen. Furthermore, we discovered that haploid loss of Gata3 in p18 deficient mice led to the development of B cell lymphomas that were capable of rapidly regenerating tumors when transplanted into immunocompromised mice. These results indicate that Gata3 deficiency promotes B cell differentiation and proliferation, and cooperates with p18 loss to induce B cell lymphomas. This study, for the first time, reveals that Gata3 is a tumor suppressor specifically in B cell lymphomagenesis.
Collapse
Affiliation(s)
- Shiqin Liu
- Department of Hematology, Peking University First Hospital, Beijing, 100034, China.,Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami
| | - Ho Lam Chan
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami
| | - Feng Bai
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami
| | - Jinshan Ma
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami.,Xinjiang Uigur Autonomous Region People's Hospital, Xinjiang, 830001, China
| | - Alexandria Scott
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami.,The Sheila and David Fuente Graduate Program in Cancer Biology, Miller School of Medicine, University of Miami, FL 33136, Miami
| | - David J Robbins
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami.,Sylvester Cancer Center, Miller School of Medicine, University of Miami, FL 33136, Miami
| | - Anthony J Capobianco
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami.,Sylvester Cancer Center, Miller School of Medicine, University of Miami, FL 33136, Miami
| | - Ping Zhu
- Department of Hematology, Peking University First Hospital, Beijing, 100034, China
| | - Xin-Hai Pei
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami.,The Sheila and David Fuente Graduate Program in Cancer Biology, Miller School of Medicine, University of Miami, FL 33136, Miami.,Sylvester Cancer Center, Miller School of Medicine, University of Miami, FL 33136, Miami
| |
Collapse
|
27
|
Sau A, Cabrita MA, Pratt MAC. NF-κB at the Crossroads of Normal Mammary Gland Biology and the Pathogenesis and Prevention of BRCA1-Mutated Breast Cancer. Cancer Prev Res (Phila) 2017; 11:69-80. [PMID: 29101208 DOI: 10.1158/1940-6207.capr-17-0225] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/03/2017] [Accepted: 10/27/2017] [Indexed: 11/16/2022]
Abstract
Recent studies have shown that progesterone receptor (PR)-expressing cells respond to progesterone in part through the induction of the receptor activator of NF-κB ligand (RANKL), which acts in a paracrine manner to induce expansion of a RANK-expressing luminal progenitor cell population. The RANK+ population in human breast tissue from carriers of BRCA1 mutations (BRCA1mut/+) as well as the luminal progenitor population in Brca1-deficient mouse mammary glands is abnormally amplified. Remarkably, mouse Brca1+/- and human BRCA1mut/+ progenitor cells are able to form colonies in vitro in the absence of progesterone, demonstrating a hormone-independent proliferative capacity. Our research has demonstrated that proliferation in BRCA1-deficient cells results in a DNA damage response (DDR) that activates a persistent NF-κB signal, which supplants progesterone/RANKL signaling for an extended time period. Thus, the transcriptional targets normally activated by RANKL that promote a proliferative response in luminal progenitors can contribute to the susceptibility of mammary epithelial cells to BRCA1-mutated breast cancers as a consequence of DDR-induced NF-κB. Together, these latest findings mark substantial progress in uncovering the mechanisms driving high rates of breast tumorigenesis in BRCA1 mutation carriers and ultimately reveal possibilities for nonsurgical prevention strategies. Cancer Prev Res; 11(2); 69-80. ©2017 AACR.
Collapse
Affiliation(s)
- Andrea Sau
- University of Ottawa, Ottawa, Ontario, Canada
| | | | | |
Collapse
|
28
|
Scott A, Bai F, Chan HL, Liu S, Slingerland JM, Robbins DJ, Capobianco AJ, Pei XH. p16 loss rescues functional decline of Brca1-deficient mammary stem cells. Cell Cycle 2017; 16:759-764. [PMID: 28278054 PMCID: PMC5405722 DOI: 10.1080/15384101.2017.1295185] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 01/19/2017] [Accepted: 02/08/2017] [Indexed: 01/04/2023] Open
Abstract
Recent evidence indicates that the accumulation of endogenous DNA damage can induce senescence and limit the function of adult stem cells. It remains elusive whether deficiency in DNA damage repair is associated with the functional alteration of mammary stem cells. In this article, we reported that senescence was induced in mammary epithelial cells during aging along with increased expression of p16Ink4a (p16), an inhibitor of CDK4 and CKD6. Loss of p16 abrogated the age-induced senescence in mammary epithelial cells and significantly increased mammary stem cell function. We showed that loss of Brca1, a tumor suppressor that functions in DNA damage repair, in the mammary epithelium induced senescence with induction of p16 and a decline of stem cell function, which was rescued by p16 loss. These data not only answer the question as to whether deficiency in DNA damage repair is associated with the functional decline of mammary stem cells, but also identify the role of p16 in suppressing Brca1-deficient mammary stem cell function.
Collapse
Affiliation(s)
- Alexandria Scott
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- The Sheila and David Fuente Graduate Program in Cancer Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Feng Bai
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Ho Lam Chan
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Shiqin Liu
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Joyce M. Slingerland
- Braman Family Breast Cancer Institute, Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - David J. Robbins
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Anthony J. Capobianco
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Xin-Hai Pei
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- The Sheila and David Fuente Graduate Program in Cancer Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
29
|
Mammary stem cells: angels or demons in mammary gland? Signal Transduct Target Ther 2017; 2:16038. [PMID: 29263909 PMCID: PMC5661614 DOI: 10.1038/sigtrans.2016.38] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 12/04/2016] [Accepted: 12/07/2016] [Indexed: 02/08/2023] Open
Abstract
A highly dynamic development process exits within the epithelia of mammary gland, featuring morphogenetic variation during puberty, pregnancy, lactation, and regression. The identification of mammary stem cells (MaSCs) via lineage-tracing studies has substantiated a hierarchical organization of the mammary epithelia. A single MaSC is capable of reconstituting the entirely functional mammary gland upon orthotopic transplantation. Although different mammary cell subpopulations can be candidate cells-of-origin for distinct breast tumor subtypes, it still lacks experimental proofs whether MaSCs, the most primitive cells, are the ‘seeds’ of malignant transformation during most, if not all, tumorigenesis in the breast. Here, we review current knowledge of mammary epithelial hierarchy, highlighting the roles of mammary stem/progenitor cells and breast cancer stem cells (BCSCs) along with their key molecular regulators in organ development and cancer evolution. Clarifying these issues will pave the way for developing novel interventions toward stem/progenitor cells in either prevention or treatment of breast cancer (BrCa).
Collapse
|
30
|
McBryan J, Howlin J. Pubertal Mammary Gland Development: Elucidation of In Vivo Morphogenesis Using Murine Models. Methods Mol Biol 2017; 1501:77-114. [PMID: 27796948 DOI: 10.1007/978-1-4939-6475-8_3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
During the past 25 years, the combination of increasingly sophisticated gene targeting technology with transplantation techniques has allowed researchers to address a wide array of questions about postnatal mammary gland development. These in turn have significantly contributed to our knowledge of other branched epithelial structures. This review chapter highlights a selection of the mouse models exhibiting a pubertal mammary gland phenotype with a focus on how they have contributed to our overall understanding of in vivo mammary morphogenesis. We discuss mouse models that have enabled us to assign functions to particular genes and proteins and, more importantly, have determined when and where these factors are required for completion of ductal outgrowth and branch patterning. The reason for the success of the mouse mammary gland model is undoubtedly the suitability of the postnatal mammary gland to experimental manipulation. The gland itself is very amenable to investigation and the combination of genetic modification with accessibility to the tissue has allowed an impressive number of studies to inform biology. Excision of the rudimentary epithelial structure postnatally allows genetically modified tissue to be readily transplanted into wild type stroma or vice versa, and has thus defined the contribution of each compartment to particular phenotypes. Similarly, whole gland transplantation has been used to definitively discern local effects from indirect systemic effects of various growth factors and hormones. While appreciative of the power of these tools and techniques, we are also cognizant of some of their limitations, and we discuss some shortcomings and future strategies that can overcome them.
Collapse
Affiliation(s)
- Jean McBryan
- Department of Molecular Medicine Royal College of Surgeons in Ireland Education and Research Centre, Beaumont Hospital, Dublin, 9, Ireland
| | - Jillian Howlin
- Division of Oncology-Pathology, Lund University Cancer Center/Medicon Village, Building 404:B2, Scheelevägen 2, 223 81, Lund, Sweden.
| |
Collapse
|
31
|
Scott A, Bai F, Chan HL, Liu S, Ma J, Slingerland JM, Robbins DJ, Capobianco AJ, Pei XH. p16INK4a suppresses BRCA1-deficient mammary tumorigenesis. Oncotarget 2016; 7:84496-84507. [PMID: 27811360 PMCID: PMC5356676 DOI: 10.18632/oncotarget.13015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/25/2016] [Indexed: 11/25/2022] Open
Abstract
Senescence prevents the proliferation of genomically damaged, but otherwise replication competent cells at risk of neoplastic transformation. p16INK4A (p16), an inhibitor of CDK4 and CDK6, plays a critical role in controlling cellular senescence in multiple organs. Functional inactivation of p16 by gene mutation and promoter methylation is frequently detected in human breast cancers. However, deleting p16 in mice or targeting DNA methylation within the murine p16 promoter does not result in mammary tumorigenesis. How loss of p16 contributes to mammary tumorigenesis in vivo is not fully understood.In this article, we reported that disruption of Brca1 in the mammary epithelium resulted in premature senescence that was rescued by p16 loss. We found that p16 loss transformed Brca1-deficient mammary epithelial cells and induced mammary tumors, though p16 loss alone was not sufficient to induce mammary tumorigenesis. We demonstrated that loss of both p16 and Brca1 led to metastatic, basal-like, mammary tumors with the induction of EMT and an enrichment of tumor initiating cells. We discovered that promoter methylation silenced p16 expression in most of the tumors developed in mice heterozygous for p16 and lacking Brca1. These data not only identified the function of p16 in suppressing BRCA1-deficient mammary tumorigenesis, but also revealed a collaborative effect of genetic mutation of p16 and epigenetic silencing of its transcription in promoting tumorigenesis. To the best of our knowledge, this is the first genetic evidence directly showing that p16 which is frequently deleted and inactivated in human breast cancers, collaborates with Brca1 controlling mammary tumorigenesis.
Collapse
MESH Headings
- Animals
- BRCA1 Protein/genetics
- BRCA1 Protein/metabolism
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cells, Cultured
- Cyclin-Dependent Kinase Inhibitor p16/genetics
- Cyclin-Dependent Kinase Inhibitor p16/metabolism
- DNA Methylation
- Epithelial Cells/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/metabolism
- Mammary Neoplasms, Animal/pathology
- Mice, Knockout
- Mice, Transgenic
- Promoter Regions, Genetic/genetics
Collapse
Affiliation(s)
- Alexandria Scott
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- The Sheila and David Fuente Graduate Program in Cancer Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Feng Bai
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Ho Lam Chan
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Shiqin Liu
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Jinshan Ma
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Joyce M Slingerland
- Braman Family Breast Cancer Institute, Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - David J. Robbins
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Anthony J. Capobianco
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Xin-Hai Pei
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- The Sheila and David Fuente Graduate Program in Cancer Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
32
|
Incidence of BRCA1 somatic mutations and response to neoadjuvant chemotherapy in Chinese women with triple-negative breast cancer. Gene 2016; 584:26-30. [PMID: 26956035 DOI: 10.1016/j.gene.2016.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 02/25/2016] [Accepted: 03/02/2016] [Indexed: 01/27/2023]
Abstract
BACKGROUND The prevalence of BRCA1 somatic mutations status in triple-negative breast cancer (TNBC) has not been well documented. The aims of this study were to determine the frequency of BRCA1 somatic mutations and to investigate the association between BRCA1 deleterious somatic mutation status and response to neoadjuvant chemotherapy in women with TNBC. METHODS Two hundred and five TNBC patients without BRCA1 germline mutations were enrolled in this study. Fresh tumor tissues were available for this cohort of 205 patients, including 112 patients with fresh core needle biopsy tumor tissues before treatment and 93 patients with fresh tumor tissues procured after surgery. BRCA1 somatic mutations were determined in the tumor samples using PCR-direct sequencing assay. Among the 112 patients with core needle biopsy samples, 97 patients received neoadjuvant chemotherapy. RESULTS Eight patients (3.9%) carried a BRCA1 pathogenic somatic mutation in this cohort of 205 TNBC patients. These eight BRCA1 deleterious somatic mutations included five frameshift or nonsense mutations (c.191_212del22, c.1664delA, c.4674_4675+17del, c.3671_3672insTTCC, c.1162A>T), one splicing site mutation (c.134+2T>G) and two missense mutations (c.5511G>C and c.286G>A). No significant differences in tumor characteristics between BRCA1 deleterious somatic mutation carriers and non-carriers were observed. The pCR (pathologic complete response) rate was 32.0% in the 97 patients who received neoadjuvant chemotherapy. BRCA1 deleterious somatic mutation carriers (n=5) had a higher pCR rate than did non-carriers (n=92) (BRCA1 carriers vs non-carriers, 60.0% vs 30.4%, P=0.32), although it did not reach a significance due to a small sample size. CONCLUSIONS A small subset of TNBC patients carried a BRCA1 deleterious somatic mutation; BRCA1 somatic mutation carriers are likely to respond to neoadjuvant chemotherapy.
Collapse
|
33
|
Watson JK, Rulands S, Wilkinson AC, Wuidart A, Ousset M, Van Keymeulen A, Göttgens B, Blanpain C, Simons BD, Rawlins EL. Clonal Dynamics Reveal Two Distinct Populations of Basal Cells in Slow-Turnover Airway Epithelium. Cell Rep 2015; 12:90-101. [PMID: 26119728 PMCID: PMC4518462 DOI: 10.1016/j.celrep.2015.06.011] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 04/25/2015] [Accepted: 06/02/2015] [Indexed: 01/06/2023] Open
Abstract
Epithelial lineages have been studied at cellular resolution in multiple organs that turn over rapidly. However, many epithelia, including those of the lung, liver, pancreas, and prostate, turn over slowly and may be regulated differently. We investigated the mouse tracheal epithelial lineage at homeostasis by using long-term clonal analysis and mathematical modeling. This pseudostratified epithelium contains basal cells and secretory and multiciliated luminal cells. Our analysis revealed that basal cells are heterogeneous, comprising approximately equal numbers of multipotent stem cells and committed precursors, which persist in the basal layer for 11 days before differentiating to luminal fate. We confirmed the molecular and functional differences within the basal population by using single-cell qRT-PCR and further lineage labeling. Additionally, we show that self-renewal of short-lived secretory cells is a feature of homeostasis. We have thus revealed early luminal commitment of cells that are morphologically indistinguishable from stem cells.
Collapse
Affiliation(s)
- Julie K Watson
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Wellcome Trust-MRC Stem Cell Institute University of Cambridge, Cambridge CB2 3EG, UK; Department of Pathology, University of Cambridge, Cambridge CB2 3EG, UK
| | - Steffen Rulands
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Wellcome Trust-MRC Stem Cell Institute University of Cambridge, Cambridge CB2 3EG, UK; Cavendish Laboratory, Department of Physics, J. J. Thomson Avenue, Cambridge CB3 0HE, UK
| | - Adam C Wilkinson
- Wellcome Trust-MRC Stem Cell Institute University of Cambridge, Cambridge CB2 3EG, UK; Department of Haematology, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK; Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK
| | - Aline Wuidart
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Marielle Ousset
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Alexandra Van Keymeulen
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Berthold Göttgens
- Wellcome Trust-MRC Stem Cell Institute University of Cambridge, Cambridge CB2 3EG, UK; Department of Haematology, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK; Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK
| | - Cédric Blanpain
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles, Brussels 1070, Belgium; Welbio, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Benjamin D Simons
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Wellcome Trust-MRC Stem Cell Institute University of Cambridge, Cambridge CB2 3EG, UK; Cavendish Laboratory, Department of Physics, J. J. Thomson Avenue, Cambridge CB3 0HE, UK
| | - Emma L Rawlins
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Wellcome Trust-MRC Stem Cell Institute University of Cambridge, Cambridge CB2 3EG, UK; Department of Pathology, University of Cambridge, Cambridge CB2 3EG, UK.
| |
Collapse
|
34
|
Seoane S, Arias E, Sigueiro R, Sendon-Lago J, Martinez-Ordoñez A, Castelao E, Eiró N, Garcia-Caballero T, Macia M, Lopez-Lopez R, Maestro M, Vizoso F, Mouriño A, Perez-Fernandez R. Pit-1 inhibits BRCA1 and sensitizes human breast tumors to cisplatin and vitamin D treatment. Oncotarget 2015; 6:14456-71. [PMID: 25992773 PMCID: PMC4546479 DOI: 10.18632/oncotarget.3894] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/25/2015] [Indexed: 11/25/2022] Open
Abstract
The POU class 1 homeobox 1 (POU1F1, also known as Pit-1), pertaining to the Pit-Oct-Unc (POU) family of transcription factors, has been related to tumor growth and metastasis in breast. However, its role in response to breast cancer therapy is unknown. We found that Pit-1 down-regulated DNA-damage and repair genes, and specifically inhibited BRCA1 gene expression, sensitizing breast cancer cells to DNA-damage agents. Administration of 1α, 25-dihydroxy-3-epi-vitamin D3 (3-Epi, an endogenous low calcemic vitamin D metabolite) reduced Pit-1 expression, and synergized with cisplatin, thus, decreasing cell proliferation and apoptosis in vitro, and reducing tumor growth in vivo. In addition, fifteen primary cultures of human breast tumors showed significantly decreased proliferation when treated with 3-Epi+cisplatin, compared to cisplatin alone. This response positively correlated with Pit-1 levels. Our findings demonstrate that high levels of Pit-1 and reduced BRCA1 levels increase breast cancer cell susceptibility to 3-Epi+cisplatin therapy.
Collapse
Affiliation(s)
- Samuel Seoane
- Department of Physiology, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
- Center for Research in Molecular Medicine and Chronic Diseases-CIMUS, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Efigenia Arias
- Center for Research in Molecular Medicine and Chronic Diseases-CIMUS, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
- Department of Obstetrics and Gynecology, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Rita Sigueiro
- Department Organic Chemistry, Research Laboratory Ignacio Rivas, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Juan Sendon-Lago
- Department of Physiology, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
- Center for Research in Molecular Medicine and Chronic Diseases-CIMUS, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Anxo Martinez-Ordoñez
- Department of Physiology, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
- Center for Research in Molecular Medicine and Chronic Diseases-CIMUS, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Esteban Castelao
- Oncology and Genetics Unit, Biomedical Research Institute of Vigo (IBIV), Complejo Hospitalario Universitario de Vigo, Servicio Galego de Saude (SERGAS), Vigo 36036, Spain
| | - Noemí Eiró
- Research Unit, Fundación Hospital de Jove, Gijón 33290, Spain
| | - Tomás Garcia-Caballero
- Department of Morphological Sciences, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Manuel Macia
- Department of Obstetrics and Gynecology, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Rafael Lopez-Lopez
- Department of Clinical Oncology, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Miguel Maestro
- Department Organic Chemistry, Research Laboratory Ignacio Rivas, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | | | - Antonio Mouriño
- Department Organic Chemistry, Research Laboratory Ignacio Rivas, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Roman Perez-Fernandez
- Department of Physiology, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
- Center for Research in Molecular Medicine and Chronic Diseases-CIMUS, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| |
Collapse
|
35
|
Cyr AR, Kulak MV, Park JM, Bogachek MV, Spanheimer PM, Woodfield GW, White-Baer LS, O’Malley YQ, Sugg SL, Olivier AK, Zhang W, Domann FE, Weigel RJ. TFAP2C governs the luminal epithelial phenotype in mammary development and carcinogenesis. Oncogene 2015; 34:436-44. [PMID: 24469049 PMCID: PMC4112181 DOI: 10.1038/onc.2013.569] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 11/12/2013] [Accepted: 12/03/2013] [Indexed: 02/07/2023]
Abstract
Molecular subtypes of breast cancer are characterized by distinct patterns of gene expression that are predictive of outcome and response to therapy. The luminal breast cancer subtypes are defined by the expression of estrogen receptor-alpha (ERα)-associated genes, many of which are directly responsive to the transcription factor activator protein 2C (TFAP2C). TFAP2C participates in a gene regulatory network controlling cell growth and differentiation during ectodermal development and regulating ESR1/ERα and other luminal cell-associated genes in breast cancer. TFAP2C has been established as a prognostic factor in human breast cancer, however, its role in the establishment and maintenance of the luminal cell phenotype during carcinogenesis and mammary gland development have remained elusive. Herein, we demonstrate a critical role for TFAP2C in maintaining the luminal phenotype in human breast cancer and in influencing the luminal cell phenotype during normal mammary development. Knockdown of TFAP2C in luminal breast carcinoma cells induced epithelial-mesenchymal transition with morphological and phenotypic changes characterized by a loss of luminal-associated gene expression and a concomitant gain of basal-associated gene expression. Conditional knockout of the mouse homolog of TFAP2C, Tcfap2c, in mouse mammary epithelium driven by MMTV-Cre promoted aberrant growth of the mammary tree leading to a reduction in the CD24(hi)/CD49f(mid) luminal cell population and concomitant gain of the CD24(mid)/CD49f(hi) basal cell population at maturity. Our results establish TFAP2C as a key transcriptional regulator for maintaining the luminal phenotype in human breast carcinoma. Furthermore, Tcfap2c influences development of the luminal cell type during mammary development. The data suggest that TFAP2C has an important role in regulated luminal-specific genes and may be a viable therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Anthony R Cyr
- Department of Surgery, University of Iowa, Iowa City, IA, USA
- Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
| | | | - Jung M. Park
- Department of Surgery, University of Iowa, Iowa City, IA, USA
| | | | | | | | | | | | - Sonia L. Sugg
- Department of Surgery, University of Iowa, Iowa City, IA, USA
| | | | - Weizhou Zhang
- Department of Pathology, University of Iowa, Iowa City, IA, USA
| | - Frederick E. Domann
- Department of Surgery, University of Iowa, Iowa City, IA, USA
- Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
- Department of Pathology, University of Iowa, Iowa City, IA, USA
| | - Ronald J. Weigel
- Department of Surgery, University of Iowa, Iowa City, IA, USA
- Department of Biochemistry, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
36
|
Bai F, Chan HL, Scott A, Smith MD, Fan C, Herschkowitz JI, Perou CM, Livingstone AS, Robbins DJ, Capobianco AJ, Pei XH. BRCA1 suppresses epithelial-to-mesenchymal transition and stem cell dedifferentiation during mammary and tumor development. Cancer Res 2014; 74:6161-72. [PMID: 25239453 DOI: 10.1158/0008-5472.can-14-1119] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BRCA1 mutation carriers are predisposed to developing basal-like breast cancers with high metastasis and poor prognosis. Yet, how BRCA1 suppresses formation of basal-like breast cancers is still obscure. Deletion of p18(Ink4c) (p18), an inhibitor of CDK4 and CDK6, functionally inactivates the RB pathway, stimulates mammary luminal stem cell (LSC) proliferation, and leads to spontaneous luminal tumor development. Alternately, germline mutation of Brca1 shifts the fate of luminal cells to cause luminal-to-basal mammary tumor transformation. Here, we report that disrupting Brca1 by either germline or epithelium-specific mutation in p18-deficient mice activates epithelial-to-mesenchymal transition (EMT) and induces dedifferentiation of LSCs, which associate closely with expansion of basal and cancer stem cells and formation of basal-like tumors. Mechanistically, BRCA1 bound to the TWIST promoter, suppressing its activity and inhibiting EMT in mammary tumor cells. In human luminal cancer cells, BRCA1 silencing was sufficient to activate TWIST and EMT and increase tumor formation. In parallel, TWIST expression and EMT features correlated inversely with BRCA1 expression in human breast cancers. Together, our findings showed that BRCA1 suppressed TWIST and EMT, inhibited LSC dedifferentiation, and repressed expansion of basal stem cells and basal-like tumors. Thus, our work offers the first genetic evidence that Brca1 directly suppresses EMT and LSC dedifferentiation during breast tumorigenesis.
Collapse
Affiliation(s)
- Feng Bai
- Molecular Oncology Program, Department of Surgery and
| | - Ho Lam Chan
- Molecular Oncology Program, Department of Surgery and
| | | | - Matthew D Smith
- Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Cheng Fan
- Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jason I Herschkowitz
- Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Charles M Perou
- Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | - David J Robbins
- Molecular Oncology Program, Department of Surgery and Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Anthony J Capobianco
- Molecular Oncology Program, Department of Surgery and Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Xin-Hai Pei
- Molecular Oncology Program, Department of Surgery and Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida.
| |
Collapse
|
37
|
Abstract
Based on transplantation and lineage tracing studies, a hierarchy of stem and progenitor cells has been shown to exist among the mammary epithelium. In this review, Visvader and Stingl integrate recent data on the mammary stem cell differentiation hierarchy and its control at the transcriptional and epigenetic levels. They also discuss the relevance of the evolving hierarchy to the identification of “cells of origin” of breast cancer. The mammary epithelium is highly responsive to local and systemic signals, which orchestrate morphogenesis of the ductal tree during puberty and pregnancy. Based on transplantation and lineage tracing studies, a hierarchy of stem and progenitor cells has been shown to exist among the mammary epithelium. Lineage tracing has highlighted the existence of bipotent mammary stem cells (MaSCs) in situ as well as long-lived unipotent cells that drive morphogenesis and homeostasis of the ductal tree. Moreover, there is accumulating evidence for a heterogeneous MaSC compartment comprising fetal MaSCs, slow-cycling cells, and both long-term and short-term repopulating cells. In parallel, diverse luminal progenitor subtypes have been identified in mouse and human mammary tissue. Elucidation of the normal cellular hierarchy is an important step toward understanding the “cells of origin” and molecular perturbations that drive breast cancer.
Collapse
Affiliation(s)
- Jane E Visvader
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville VIC 3010, Australia
| | - John Stingl
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| |
Collapse
|
38
|
Bogachek MV, Chen Y, Kulak MV, Woodfield GW, Cyr AR, Park JM, Spanheimer PM, Li Y, Li T, Weigel RJ. Sumoylation pathway is required to maintain the basal breast cancer subtype. Cancer Cell 2014; 25:748-61. [PMID: 24835590 PMCID: PMC4096794 DOI: 10.1016/j.ccr.2014.04.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 11/12/2013] [Accepted: 04/11/2014] [Indexed: 01/28/2023]
Abstract
The TFAP2C/AP-2γ transcription factor regulates luminal breast cancer genes, and loss of TFAP2C induces epithelial-mesenchymal transition. By contrast, the highly homologous family member, TFAP2A, lacks transcriptional activity at luminal gene promoters. A detailed structure-function analysis identified that sumoylation of TFAP2A blocks its ability to induce the expression of luminal genes. Disruption of the sumoylation pathway by knockdown of sumoylation enzymes, mutation of the SUMO-target lysine of TFAP2A, or treatment with sumoylation inhibitors induced a basal-to-luminal transition, which was dependent on TFAP2A. Sumoylation inhibitors cleared the CD44(+/hi)/CD24(-/low) cell population characterizing basal cancers and inhibited tumor outgrowth of basal cancer xenografts. These findings establish a critical role for sumoylation in regulating the transcriptional mechanisms that maintain the basal cancer phenotype.
Collapse
Affiliation(s)
- Maria V Bogachek
- Department of Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Yizhen Chen
- Department of Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Mikhail V Kulak
- Department of Surgery, University of Iowa, Iowa City, IA 52242, USA
| | | | - Anthony R Cyr
- Department of Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Jung M Park
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | | | - Yingyue Li
- Department of Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Tiandao Li
- Department of Surgery, University of Iowa, Iowa City, IA 52242, USA; The Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Ronald J Weigel
- Department of Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA; Department of Biochemistry, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
39
|
|
40
|
Smits BMG, Haag JD, Rissman AI, Sharma D, Tran A, Schoenborn AA, Baird RC, Peiffer DS, Leinweber DQ, Muelbl MJ, Meilahn AL, Eichelberg MR, Leng N, Kendziorski C, John MC, Powers PA, Alexander CM, Gould MN. The gene desert mammary carcinoma susceptibility locus Mcs1a regulates Nr2f1 modifying mammary epithelial cell differentiation and proliferation. PLoS Genet 2013; 9:e1003549. [PMID: 23785296 PMCID: PMC3681674 DOI: 10.1371/journal.pgen.1003549] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 04/23/2013] [Indexed: 12/28/2022] Open
Abstract
Genome-wide association studies have revealed that many low-penetrance breast cancer susceptibility loci are located in non-protein coding genomic regions; however, few have been characterized. In a comparative genetics approach to model such loci in a rat breast cancer model, we previously identified the mammary carcinoma susceptibility locus Mcs1a. We now localize Mcs1a to a critical interval (277 Kb) within a gene desert. Mcs1a reduces mammary carcinoma multiplicity by 50% and acts in a mammary cell-autonomous manner. We developed a megadeletion mouse model, which lacks 535 Kb of sequence containing the Mcs1a ortholog. Global gene expression analysis by RNA-seq revealed that in the mouse mammary gland, the orphan nuclear receptor gene Nr2f1/Coup-tf1 is regulated by Mcs1a. In resistant Mcs1a congenic rats, as compared with susceptible congenic control rats, we found Nr2f1 transcript levels to be elevated in mammary gland, epithelial cells, and carcinoma samples. Chromatin looping over ∼820 Kb of sequence from the Nr2f1 promoter to a strongly conserved element within the Mcs1a critical interval was identified. This element contains a 14 bp indel polymorphism that affects a human-rat-mouse conserved COUP-TF binding motif and is a functional Mcs1a candidate. In both the rat and mouse models, higher Nr2f1 transcript levels are associated with higher abundance of luminal mammary epithelial cells. In both the mouse mammary gland and a human breast cancer global gene expression data set, we found Nr2f1 transcript levels to be strongly anti-correlated to a gene cluster enriched in cell cycle-related genes. We queried 12 large publicly available human breast cancer gene expression studies and found that the median NR2F1 transcript level is consistently lower in 'triple-negative' (ER-PR-HER2-) breast cancers as compared with 'receptor-positive' breast cancers. Our data suggest that the non-protein coding locus Mcs1a regulates Nr2f1, which is a candidate modifier of differentiation, proliferation, and mammary cancer risk.
Collapse
Affiliation(s)
- Bart M. G. Smits
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Jill D. Haag
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Anna I. Rissman
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Deepak Sharma
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Ann Tran
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Alexi A. Schoenborn
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Rachael C. Baird
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Dan S. Peiffer
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - David Q. Leinweber
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Matthew J. Muelbl
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Amanda L. Meilahn
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Mark R. Eichelberg
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Ning Leng
- Department of Statistics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Christina Kendziorski
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Manorama C. John
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Patricia A. Powers
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Caroline M. Alexander
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Michael N. Gould
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
41
|
Ng T, Irshad S, Stebbing J. BRCA1 mutations and luminal-basal transformation. Oncogene 2013; 32:2712-4. [PMID: 22926516 DOI: 10.1038/onc.2012.379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 07/02/2012] [Accepted: 07/04/2012] [Indexed: 12/14/2022]
Abstract
The multifunctional roles of BRCA1 include its ability to regulate transcriptional processes that control differentiation at multiple levels, as well as functioning as a tumor suppressor. Data herein demonstrate that germline mutations in Brca1 impair luminal cell lineage and mammary development, with its deficiency converting ER-positive luminal tumors into basal-like cancers. Heterozygous mutations in Brca1 lead to downregulation of a number of luminal differentiation genes, explaining how it suppresses basal-like tumors, also highlighting its importance outside of its known highly publicized role in DNA repair.
Collapse
Affiliation(s)
- T Ng
- Richard Dimbleby Department of Cancer Research, Randall Division and Division of Cancer Studies, Kings College London, Guy's Medical School Campus, London, UK
| | | | | |
Collapse
|