1
|
Bonadeo N, Chimento A, Mejía ME, Dallard BE, Sorianello E, Becu-Villalobos D, Lacau-Mengido I, Cristina C. NOTCH and IGF1 signaling systems are involved in the effects exerted by anthelminthic treatment of heifers on the bovine mammary gland. Vet Parasitol 2025; 334:110390. [PMID: 39798247 DOI: 10.1016/j.vetpar.2025.110390] [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: 09/25/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
Dairy heifers with gastrointestinal nematodes have reduced growth rates, and delayed age at puberty and milk production onset related to late mammary gland development. IGF1 and Notch signaling systems are important in this process, and an altered profile of serum IGF1 has been associated with the detrimental effect of the nematodes on parenchymal development. In this context, we aimed to study the molecular mechanisms involved in bovine mammary gland development around pre and postpuberty, focusing on proliferative and angiogenic processes that involve the Notch and IGF1 pathways. We used mammary tissue samples from pre and pubertal heifers, treated or untreated with anthelmintics, and MAC-T bovine mammary epithelial cells in vitro. Anthelminthic treatment effectively lowered EPG in feces. Mammary glands from treated heifers had increased proliferation rate (measured by PCNA) and angiogenic marker expression (VEGF and CD34), as well as increased αSMA area compared to age-matched control parasitized heifers. These changes were preceded by increased expression of Notch targets at 20 wk of age (HES1, HEY2, and HEY1), indicating a possible interaction. Similarly, IGF1R expression was increased at 30 weeks of age. To study the crosstalk between systems, bovine MAC-T cells were treated with DAPT (50 μM) to inhibit Notch signaling. DAPT decreased the proliferation of cells as evidenced by a decrease in PCNA, pERK, CYCYLIN D1; and the wound healing capacity of HMEC cells was impaired in the presence of the supernatants of DAPT-treated cells. Furthermore, DAPT decreased IGF1 and increased IGF1R mRNA levels in MAC-T cells. On the other hand, cells treated with 10 ng/mL IGF1 Increased their proliferation (MTS assay), and induced a strong tendency to increase Notch target genes (HEY1, and HES1). Furthermore, IGF1 treatment tampered the decrease in the proliferation rate induced by DAPT. Finally, a positive correlation between the IGF1R and Notch target genes (HEY1, and HES1) further suggested a relation between these two signaling systems in the bovine mammary gland. In conclusion, pubertal delay related to parasitosis is counteracted by anthelminthic treatments, which increase serum IGF1, mammary cell proliferation, and angiogenesis. We postulate the Notch pathway, mainly through the HEY1 target gene, which is modulated by the IGF1 system, may regulate both proliferative and angiogenic processes favoring normal development of the bovine mammary gland during puberty. In addition, we demonstrate that the interaction between the Notch and the IGF1 pathways may affect cell proliferation.
Collapse
Affiliation(s)
- Nadia Bonadeo
- Centro de Investigaciones Básicas y Aplicadas, Universidad Nacional del Noroeste de la Provincia de Buenos Aires, Junín, Buenos Aires 6000, Argentina; Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires, CITNOBA, UNNOBA - UNSAdA - CONICET, Monteagudo 2772, Pergamino, Buenos Aires 2700, Argentina
| | - Agustina Chimento
- Centro de Investigaciones Básicas y Aplicadas, Universidad Nacional del Noroeste de la Provincia de Buenos Aires, Junín, Buenos Aires 6000, Argentina; Comisión de Investigaciones Científicas, CIC, La Plata, Buenos Aires 1900, Argentina
| | - Miguel E Mejía
- Instituto de Biología y Medicina Experimental, IBYME-CONICET, Vuelta de Obligado 2490, CABA 1428, Argentina
| | - Bibiana E Dallard
- Instituto de Ciencias Veterinarias del Litoral (ICIVET-Litoral), Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Científicas y Tecnológicas, (UNL-CONICET), Argentina; Laboratorio de Biología Celular y Molecular Aplicada, Facultad de Ciencias Veterinarias, Universidad Nacional del Litoral, Argentina, Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Cientí∼ficas y Tecnoló∼gicas, (UNL-CONICET), Argentina
| | - Eleonora Sorianello
- Instituto de Biología y Medicina Experimental, IBYME-CONICET, Vuelta de Obligado 2490, CABA 1428, Argentina
| | - Damasia Becu-Villalobos
- Instituto de Biología y Medicina Experimental, IBYME-CONICET, Vuelta de Obligado 2490, CABA 1428, Argentina
| | - Isabel Lacau-Mengido
- Instituto de Biología y Medicina Experimental, IBYME-CONICET, Vuelta de Obligado 2490, CABA 1428, Argentina
| | - Carolina Cristina
- Centro de Investigaciones Básicas y Aplicadas, Universidad Nacional del Noroeste de la Provincia de Buenos Aires, Junín, Buenos Aires 6000, Argentina; Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires, CITNOBA, UNNOBA - UNSAdA - CONICET, Monteagudo 2772, Pergamino, Buenos Aires 2700, Argentina.
| |
Collapse
|
2
|
Van Keymeulen A. Mechanisms of Regulation of Cell Fate in Breast Development and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:167-184. [PMID: 39821026 DOI: 10.1007/978-3-031-70875-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
This chapter focuses on the mechanisms of regulation of cell fate in breast development, occurring mainly after birth, as well as in breast cancer. First, we will review how the microenvironment of the breast, as well as external cues, plays a crucial role in mammary gland cell specification and will describe how it has been shown to reprogram non-mammary cells into mammary epithelial cells. Then we will focus on the transcription factors and master regulators which have been established to be determinant for basal (BC) and luminal cell (LC) identity, and will describe the experiments of ectopic expression or loss of function of these transcription factors which demonstrated that they were crucial for cell fate. We will also discuss how master regulators are involved in the fate choice of LCs between estrogen receptor (ER)-positive cells and ER- cells, which will give rise to alveolar cells upon pregnancy and lactation. We will describe how oncogene expression induces reprogramming and change of fate of mammary epithelial cells before tumor appearance, which could be an essential step in tumorigenesis. Finally, we will describe the involvement of master regulators of mammary epithelial cells in breast cancer.
Collapse
Affiliation(s)
- Alexandra Van Keymeulen
- Laboratory of Stem Cells and Cancer (LSCC), Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
3
|
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
|
4
|
Zeng J, Singh S, Zhou X, Jiang Y, Casarez E, Atkins KA, Janes KA, Zong H. A genetic mosaic mouse model illuminates the pre-malignant progression of basal-like breast cancer. Dis Model Mech 2023; 16:dmm050219. [PMID: 37815460 PMCID: PMC10668031 DOI: 10.1242/dmm.050219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 09/11/2023] [Indexed: 10/11/2023] Open
Abstract
Basal-like breast cancer (BLBC) is highly aggressive, and often characterized by BRCA1 and p53 deficiency. Although conventional mouse models enabled the investigation of BLBC at malignant stages, its initiation and pre-malignant progression remain understudied. Here, we leveraged a mouse genetic system known as mosaic analysis with double markers (MADM) to study BLBC initiation by generating rare GFP+Brca1, p53-deficient mammary cells alongside RFP+ wild-type sibling cells. After confirming the close resemblance of mammary tumors arising in this model to human BLBC at both transcriptomic and genomic levels, we focused our studies on the pre-malignant progression of BLBC. Initiated GFP+ mutant cells showed a stepwise pre-malignant progression trajectory from focal expansion to hyper-alveolarization and then to micro-invasion. Furthermore, despite morphological similarities to alveoli, hyper-alveolarized structures actually originate from ductal cells based on twin-spot analysis of GFP-RFP sibling cells. Finally, luminal-to-basal transition occurred exclusively in cells that have progressed to micro-invasive lesions. Our MADM model provides excellent spatiotemporal resolution to illuminate the pre-malignant progression of BLBC, and should enable future studies on early detection and prevention for this cancer.
Collapse
Affiliation(s)
- Jianhao Zeng
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Shambhavi Singh
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Xian Zhou
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Ying Jiang
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Eli Casarez
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Kristen A. Atkins
- Department of Pathology, University of Virginia Health System, Charlottesville, VA 22908, USA
- University of Virginia Comprehensive Cancer Center, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Kevin A. Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
- University of Virginia Comprehensive Cancer Center, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Hui Zong
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
- University of Virginia Comprehensive Cancer Center, University of Virginia Health System, Charlottesville, VA 22908, USA
| |
Collapse
|
5
|
Zeng J, Singh S, Jiang Y, Casarez E, Atkins KA, Janes KA, Zong H. A genetic mosaic mouse model illuminates the pre-malignant progression of basal-like breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.25.538333. [PMID: 37163037 PMCID: PMC10168298 DOI: 10.1101/2023.04.25.538333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Basal-like breast cancer is an aggressive breast cancer subtype, often characterized by a deficiency in BRCA1 function and concomitant loss of p53 . While conventional mouse models enable the investigation of its malignant stages, one that reveals its initiation and pre-malignant progression is lacking. Here, we leveraged a mouse genetic system known as M osaic A nalysis with D ouble M arkers (MADM) to generate rare GFP-labeled Brca1 , p53 -deficient cells alongside RFP+ wildtype sibling cells in the mammary gland. The mosaicism resembles the sporadic initiation of human cancer and enables spatially resolved analysis of mutant cells in comparison to paired wildtype sibling cells. Mammary tumors arising in the model show transcriptomic and genomic characteristics similar to human basal-like breast cancer. Analysis of GFP+ mutant cells at interval time points before malignancy revealed a stepwise progression of lesions from focal expansion to hyper-alveolarization and then to micro-invasion. These stereotyped morphologies indicate the pre-malignant stage irrespective of the time point at which it is observed. Paired analysis of GFP-RFP siblings during focal expansion suggested that hyper-alveolarized structures originate from ductal rather than alveolar cells, despite their morphological similarities to alveoli. Evidence for luminal-to-basal transition at the pre-malignant stages was restricted to cells that had escaped hyper-alveoli and progressed to micro-invasive lesions. Our MADM-based mouse model presents a useful tool for studying the pre-malignancy of basal-like breast cancer. Summary statement A mouse model recapitulates the process of human basal-like breast tumorigenesis initiated from sporadic Brca1, p53 -deficient cells, empowering spatially-resolved analysis of mutant cells during pre-malignant progression.
Collapse
|
6
|
Gray GK, Li CMC, Rosenbluth JM, Selfors LM, Girnius N, Lin JR, Schackmann RCJ, Goh WL, Moore K, Shapiro HK, Mei S, D'Andrea K, Nathanson KL, Sorger PK, Santagata S, Regev A, Garber JE, Dillon DA, Brugge JS. A human breast atlas integrating single-cell proteomics and transcriptomics. Dev Cell 2022; 57:1400-1420.e7. [PMID: 35617956 DOI: 10.1016/j.devcel.2022.05.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/23/2022] [Accepted: 05/02/2022] [Indexed: 12/12/2022]
Abstract
The breast is a dynamic organ whose response to physiological and pathophysiological conditions alters its disease susceptibility, yet the specific effects of these clinical variables on cell state remain poorly annotated. We present a unified, high-resolution breast atlas by integrating single-cell RNA-seq, mass cytometry, and cyclic immunofluorescence, encompassing a myriad of states. We define cell subtypes within the alveolar, hormone-sensing, and basal epithelial lineages, delineating associations of several subtypes with cancer risk factors, including age, parity, and BRCA2 germline mutation. Of particular interest is a subset of alveolar cells termed basal-luminal (BL) cells, which exhibit poor transcriptional lineage fidelity, accumulate with age, and carry a gene signature associated with basal-like breast cancer. We further utilize a medium-depletion approach to identify molecular factors regulating cell-subtype proportion in organoids. Together, these data are a rich resource to elucidate diverse mammary cell states.
Collapse
Affiliation(s)
- G Kenneth Gray
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Carman Man-Chung Li
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Jennifer M Rosenbluth
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute (DFCI), Boston, MA 02115, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Laura M Selfors
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Nomeda Girnius
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA; The Laboratory of Systems Pharmacology (LSP), HMS, Boston, MA 02115, USA
| | - Jia-Ren Lin
- The Laboratory of Systems Pharmacology (LSP), HMS, Boston, MA 02115, USA
| | - Ron C J Schackmann
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Walter L Goh
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Kaitlin Moore
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Hana K Shapiro
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Shaolin Mei
- The Laboratory of Systems Pharmacology (LSP), HMS, Boston, MA 02115, USA
| | - Kurt D'Andrea
- Department of Medicine, Division of Translation Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Katherine L Nathanson
- Department of Medicine, Division of Translation Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peter K Sorger
- The Laboratory of Systems Pharmacology (LSP), HMS, Boston, MA 02115, USA
| | - Sandro Santagata
- The Laboratory of Systems Pharmacology (LSP), HMS, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital (BWH), Boston, MA 02115, USA
| | - Aviv Regev
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Judy E Garber
- Department of Medical Oncology, Dana-Farber Cancer Institute (DFCI), Boston, MA 02115, USA
| | - Deborah A Dillon
- Department of Pathology, Brigham and Women's Hospital (BWH), Boston, MA 02115, USA
| | - Joan S Brugge
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA.
| |
Collapse
|
7
|
Chen W, Wei W, Yu L, Ye Z, Huang F, Zhang L, Hu S, Cai C. Mammary Development and Breast Cancer: a Notch Perspective. J Mammary Gland Biol Neoplasia 2021; 26:309-320. [PMID: 34374886 PMCID: PMC8566423 DOI: 10.1007/s10911-021-09496-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 07/21/2021] [Indexed: 12/16/2022] Open
Abstract
Mammary gland development primarily occurs postnatally, and this unique process is complex and regulated by systemic hormones and local growth factors. The mammary gland is also a highly dynamic organ that undergoes profound changes at puberty and during the reproductive cycle. These changes are driven by mammary stem cells (MaSCs). Breast cancer is one of the most common causes of cancer-related death in women. Cancer stem cells (CSCs) play prominent roles in tumor initiation, drug resistance, tumor recurrence, and metastasis. The highly conserved Notch signaling pathway functions as a key regulator of the niche mediating mammary organogenesis and breast neoplasia. In this review, we discuss mechanisms by which Notch contributes to breast carcinoma pathology and suggest potentials for therapeutic targeting of Notch in breast cancer. In summary, we provide a comprehensive overview of Notch functions in regulating MaSCs, mammary development, and breast cancer.
Collapse
Affiliation(s)
- Weizhen Chen
- Department of Orthopaedics, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Wei Wei
- Department of Orthopaedics, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Liya Yu
- Department of Orthopaedics, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Zi Ye
- Department of Orthopaedics, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Fujing Huang
- Department of Orthopaedics, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Liyan Zhang
- Department of Orthopaedics, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Shiqi Hu
- DU-ANU Joint Science College, Shandong University, Weihai, 264200, China
| | - Cheguo Cai
- Department of Orthopaedics, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
8
|
Edwards A, Brennan K. Notch Signalling in Breast Development and Cancer. Front Cell Dev Biol 2021; 9:692173. [PMID: 34295896 PMCID: PMC8290365 DOI: 10.3389/fcell.2021.692173] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/07/2021] [Indexed: 12/22/2022] Open
Abstract
The Notch signalling pathway is a highly conserved developmental signalling pathway, with vital roles in determining cell fate during embryonic development and tissue homeostasis. Aberrant Notch signalling has been implicated in many disease pathologies, including cancer. In this review, we will outline the mechanism and regulation of the Notch signalling pathway. We will also outline the role Notch signalling plays in normal mammary gland development and how Notch signalling is implicated in breast cancer tumorigenesis and progression. We will cover how Notch signalling controls several different hallmarks of cancer within epithelial cells with sections focussed on its roles in proliferation, apoptosis, invasion, and metastasis. We will provide evidence for Notch signalling in the breast cancer stem cell phenotype, which also has implications for therapy resistance and disease relapse in breast cancer patients. Finally, we will summarise the developments in therapeutic targeting of Notch signalling, and the pros and cons of this approach for the treatment of breast cancer.
Collapse
Affiliation(s)
- Abigail Edwards
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Keith Brennan
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
9
|
Fernández-Chacón M, García-González I, Mühleder S, Benedito R. Role of Notch in endothelial biology. Angiogenesis 2021; 24:237-250. [PMID: 34050878 DOI: 10.1007/s10456-021-09793-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 12/23/2022]
Abstract
The Notch signalling pathway is one of the main regulators of endothelial biology. In the last 20 years the critical function of Notch has been uncovered in the context of angiogenesis, participating in tip-stalk specification, arterial-venous differentiation, vessel stabilization, and maturation processes. Importantly, pharmacological compounds targeting distinct members of the Notch signalling pathway have been used in the clinics for cancer therapy. However, the underlying mechanisms that support the variety of outcomes triggered by Notch in apparently opposite contexts such as angiogenesis and vascular homeostasis remain unknown. In recent years, advances in -omics technologies together with mosaic analysis and high molecular, cellular and temporal resolution studies have allowed a better understanding of the mechanisms driven by the Notch signalling pathway in different endothelial contexts. In this review we will focus on the main findings that revisit the role of Notch signalling in vascular biology. We will also discuss potential future directions and technologies that will shed light on the puzzling role of Notch during endothelial growth and homeostasis. Addressing these open questions may allow the improvement and development of therapeutic strategies based on modulation of the Notch signalling pathway.
Collapse
Affiliation(s)
- Macarena Fernández-Chacón
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, Madrid, Spain
| | - Irene García-González
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Severin Mühleder
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Rui Benedito
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| |
Collapse
|
10
|
Orzechowska M, Anusewicz D, Bednarek AK. Functional Gene Expression Differentiation of the Notch Signaling Pathway in Female Reproductive Tract Tissues-A Comprehensive Review With Analysis. Front Cell Dev Biol 2021; 8:592616. [PMID: 33384996 PMCID: PMC7770115 DOI: 10.3389/fcell.2020.592616] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
The Notch pathway involves evolutionarily conserved signaling regulating the development of the female tract organs such as breast, ovary, cervix, and uterine endometrium. A great number of studies revealed Notch aberrancies in association with their carcinogenesis and disease progression, the management of which is still challenging. The present study is a comprehensive review of the available literature on Notch signaling during the normal development and carcinogenesis of the female tract organs. The review has been enriched with our analyses of the TCGA data including breast, cervical, ovarian, and endometrial carcinomas concerning the effects of Notch signaling at two levels: the core components and downstream effectors, hence filling the lack of global overview of Notch-driven carcinogenesis and disease progression. Phenotype heterogeneity regarding Notch signaling was projected in two uniform manifold approximation and projection algorithm dimensions, preceded by the principal component analysis step reducing the data burden. Additionally, overall and disease-free survival analyses were performed with the optimal cutpoint determination by Evaluate Cutpoints software to establish the character of particular Notch components in tumorigenesis. In addition to the review, we demonstrated separate models of the examined cancers of the Notch pathway and its targets, although expression profiles of all normal tissues were much more similar to each other than to its cancerous compartments. Such Notch-driven cancerous differentiation resulted in a case of opposite association with DFS and OS. As a consequence, target genes also show very distinct profiles including genes associated with cell proliferation and differentiation, energy metabolism, or the EMT. In conclusion, the observed Notch associations with the female tract malignancies resulted from differential expression of target genes. This may influence a future analysis to search for new therapeutic targets based on specific Notch pathway profiles.
Collapse
Affiliation(s)
| | - Dorota Anusewicz
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | - Andrzej K Bednarek
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
11
|
Reichrath J, Reichrath S. The Impact of Notch Signaling for Carcinogenesis and Progression of Nonmelanoma Skin Cancer: Lessons Learned from Cancer Stem Cells, Tumor Angiogenesis, and Beyond. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1287:123-154. [PMID: 33034030 DOI: 10.1007/978-3-030-55031-8_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since many decades, nonmelanoma skin cancer (NMSCs) is the most common malignancy worldwide. Basal cell carcinomas (BCC) and squamous cell carcinomas (SCC) are the major types of NMSCs, representing approximately 70% and 25% of these neoplasias, respectively. Because of their continuously rising incidence rates, NMSCs represent a constantly increasing global challenge for healthcare, although they are in most cases nonlethal and curable (e.g., by surgery). While at present, carcinogenesis of NMSC is still not fully understood, the relevance of genetic and molecular alterations in several pathways, including evolutionary highly conserved Notch signaling, has now been shown convincingly. The Notch pathway, which was first developed during evolution in metazoans and that was first discovered in fruit flies (Drosophila melanogaster), governs cell fate decisions and many other fundamental processes that are of high relevance not only for embryonic development, but also for initiation, promotion, and progression of cancer. Choosing NMSC as a model, we give in this review a brief overview on the interaction of Notch signaling with important oncogenic and tumor suppressor pathways and on its role for several hallmarks of carcinogenesis and cancer progression, including the regulation of cancer stem cells, tumor angiogenesis, and senescence.
Collapse
Affiliation(s)
- Jörg Reichrath
- Department of Dermatology, Saarland University Medical Center, Homburg, Germany.
| | - Sandra Reichrath
- Department of Dermatology, Saarland University Medical Center, Homburg, Germany.,School of Health Professions, Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
12
|
Onoyama I, Nakayama S, Shimizu H, Nakayama KI. Loss of Fbxw7 Impairs Development of and Induces Heterogeneous Tumor Formation in the Mouse Mammary Gland. Cancer Res 2020; 80:5515-5530. [PMID: 33234509 DOI: 10.1158/0008-5472.can-20-0271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 08/17/2020] [Accepted: 10/22/2020] [Indexed: 11/16/2022]
Abstract
Fbxw7 is an F-box protein that contributes to regulation of cell proliferation and cell fate determination as well as to tumor suppression in various tissues. In this study, we generated mice with mammary gland-specific ablation of Fbxw7 (Blg-Cre/Fbxw7 F/F mice) and found that most neonates born to mutant dams die soon after birth as a result of defective maternal lactation. The mammary gland of mutant dams was markedly atrophic and manifested both excessive cell proliferation and apoptosis in association with the accumulation of Notch1 and p63. Despite the hypoplastic nature of the mutant mammary gland, Blg-Cre/Fbxw7 F/F mice spontaneously developed mammary tumors that resembled basal-like carcinoma with marked intratumoral heterogeneity. Additional inactivation of Trp53 in Blg-Cre/Fbxw7 F/F mice further promoted onset and development of mammary tumors, suggesting that spontaneous mutation of Trp53 may facilitate transition of hypoplastic mammary lesions to aggressive cancer in mice lacking Fbxw7. RNA-sequencing analysis of epithelial- and mesenchymal-like cell lines from a Blg-Cre/Fbxw7 F/F mouse tumor revealed an increased mutation rate and structural alterations in the tumor and differential expression of upstream transcription factors including known targets of Fbxw7. Together, our results implicate Fbxw7 in the regulation of cell differentiation and in tumor suppression in the mammary gland. Loss of Fbxw7 increases mutation rate and chromosome instability, activates signaling pathways governed by transcription factors regulated by Fbxw7, and triggers the development of mammary tumors with prominent heterogeneity. SIGNIFICANCE: Mammary gland-specific ablation of Fbxw7 in mice results in defective gland development and spontaneous mammary tumor formation reminiscent of human basal-like carcinoma with intratumoral heterogeneity. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/24/5515/F1.large.jpg.
Collapse
Affiliation(s)
- Ichiro Onoyama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka, Fukuoka Japan
| | - Shogo Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka, Fukuoka Japan
| | - Hideyuki Shimizu
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka, Fukuoka Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka, Fukuoka Japan.
| |
Collapse
|
13
|
Centonze A, Lin S, Tika E, Sifrim A, Fioramonti M, Malfait M, Song Y, Wuidart A, Van Herck J, Dannau A, Bouvencourt G, Dubois C, Dedoncker N, Sahay A, de Maertelaer V, Siebel CW, Van Keymeulen A, Voet T, Blanpain C. Heterotypic cell-cell communication regulates glandular stem cell multipotency. Nature 2020; 584:608-613. [PMID: 32848220 DOI: 10.1038/s41586-020-2632-y] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/20/2020] [Indexed: 12/15/2022]
Abstract
Glandular epithelia, including the mammary and prostate glands, are composed of basal cells (BCs) and luminal cells (LCs)1,2. Many glandular epithelia develop from multipotent basal stem cells (BSCs) that are replaced in adult life by distinct pools of unipotent stem cells1,3-8. However, adult unipotent BSCs can reactivate multipotency under regenerative conditions and upon oncogene expression3,9-13. This suggests that an active mechanism restricts BSC multipotency under normal physiological conditions, although the nature of this mechanism is unknown. Here we show that the ablation of LCs reactivates the multipotency of BSCs from multiple epithelia both in vivo in mice and in vitro in organoids. Bulk and single-cell RNA sequencing revealed that, after LC ablation, BSCs activate a hybrid basal and luminal cell differentiation program before giving rise to LCs-reminiscent of the genetic program that regulates multipotency during embryonic development7. By predicting ligand-receptor pairs from single-cell data14, we find that TNF-which is secreted by LCs-restricts BC multipotency under normal physiological conditions. By contrast, the Notch, Wnt and EGFR pathways were activated in BSCs and their progeny after LC ablation; blocking these pathways, or stimulating the TNF pathway, inhibited regeneration-induced BC multipotency. Our study demonstrates that heterotypic communication between LCs and BCs is essential to maintain lineage fidelity in glandular epithelial stem cells.
Collapse
Affiliation(s)
- Alessia Centonze
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Shuheng Lin
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Elisavet Tika
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Alejandro Sifrim
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium.,Sanger Institute-EBI Single-Cell Genomics Centre, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Marco Fioramonti
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Milan Malfait
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Yura Song
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Aline Wuidart
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Jens Van Herck
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
| | - Anne Dannau
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Gaelle Bouvencourt
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Christine Dubois
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Nina Dedoncker
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
| | - Amar Sahay
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,BROAD Institute of Harvard and MIT, Cambridge, MA, USA
| | | | - Christian W Siebel
- Department of Discovery Oncology, Genentech, South San Francisco, CA, USA
| | | | - Thierry Voet
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium.,Sanger Institute-EBI Single-Cell Genomics Centre, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Cédric Blanpain
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles (ULB), Brussels, Belgium. .,WELBIO, Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
14
|
Dettori ML, Pazzola M, Petretto E, Vacca GM. Association Analysis between SPP1, POFUT1 and PRLR Gene Variation and Milk Yield, Composition and Coagulation Traits in Sarda Sheep. Animals (Basel) 2020; 10:E1216. [PMID: 32708940 PMCID: PMC7401589 DOI: 10.3390/ani10071216] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 01/02/2023] Open
Abstract
Many studies focus on the identification of genomic regions that undergo selective processes, where evidence of selection is revealed and positional candidate genes are identified. The aim of the research was to evaluate the association between positional candidate genes, namely secreted phosphoprotein 1 (SPP1, sheep chromosome Ovis aries OAR6, 36.651-36.658 Mb), protein O-fucosyltransferase 1 (POFUT1, OAR13, 61.006-61.027 Mb) and prolactin receptor (PRLR, OAR16, 38.969-39.028 Mb) with milk yield, composition and coagulation traits. Eight single nucleotide polymorphisms (SNPs) mapping to the three genes were genotyped in 380 Sarda dairy sheep. Statistical analysis revealed an association between SNP rs161844011 at SPP1 (chromosome position Oar_v3 OAR6:36651870, gene region exon 7) and somatic cell score, while POFUT1 SNP rs424501869 (OAR13:61007495, intron 1) was associated with curd firmness both 45 and 60 min after rennet addition (p = 0.015 and p = 0.007, respectively). SNP rs400874750 at PRLR gene (OAR16:39004070, intron 2) had a significant association with lactose content (p = 0.020), somatic cell score (p = 0.038), rennet coagulation time (p = 0.018) and curd firming time (p = 0.047). The outcome of this research confirmed predictions based on genomic studies, producing new information regarding the SPP1, POFUT1 and PRLR genes, which may be useful for future breeding schemes.
Collapse
Affiliation(s)
| | - Michele Pazzola
- Department of Veterinary Medicine, University of Sassari, via Vienna 2, 07100 Sassari, Italy; (M.L.D.); (E.P.); (G.M.V.)
| | | | | |
Collapse
|
15
|
Saboya M, Jetzt AE, Datar K, Cohick WS. Fetal Alcohol Exposure Alters Mammary Epithelial Cell Subpopulations and Promotes Tumorigenesis. Alcohol Clin Exp Res 2020; 44:831-843. [PMID: 32056248 PMCID: PMC7166183 DOI: 10.1111/acer.14308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 02/05/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Fetal alcohol exposure (FAE) increases the risk of mammary tumorigenesis in adult offspring; however, the underlying mechanism remains unknown. This study tested the hypothesis that FAE shifts the mammary epithelial cell (MEC) composition toward one that promotes tumorigenesis. METHODS Pregnant Friend Virus B NIH Jackson dams bred to MMTV-Wnt1 male mice were given ad libitum access to 5% alcohol in 0.2% saccharin solution from GD9-10 and 10% alcohol in 0.2% saccharin from GD11-GD19 or 0.2% saccharin solution from GD9-GD19. Thoracic and inguinal mammary glands from wild-type (WT) and transgenic (Tg) female offspring were harvested at 5 and 10 weeks of age and dissociated to yield a single cell suspension enriched for MECs for flow cytometry, mammosphere assay, and gene analysis. A subset of Tg offspring was followed for tumor formation. RESULTS WT glands of FAE animals exhibited a decreased basal cell population and increased luminal: basal ratio at 10 weeks of age. qRT-PCR analysis of total MECs found that Hey1 mRNA expression was increased in the WT FAE group at 10 weeks of age. In Tg glands, FAE increased the luminal progenitor cell population at 5 weeks of age but did not alter MEC composition at 10 weeks of age. Tertiary mammosphere-forming efficiency was greater in the WT glands of FAE animals at 10 weeks of age. Tumor latency was decreased in the FAE group. Flow cytometry analysis indicated that FAE females developed tumors with an increased basal cell population. CONCLUSIONS These data indicate that FAE can shift MEC subpopulations, increasing the proportion of cells that are potentially vulnerable to transformation and affecting cancer risk.
Collapse
Affiliation(s)
- Mariana Saboya
- From the, The Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ
| | - Amanda E Jetzt
- From the, The Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ
| | - Ketaki Datar
- From the, The Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ
| | - Wendie S Cohick
- From the, The Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ
| |
Collapse
|
16
|
Fu NY, Nolan E, Lindeman GJ, Visvader JE. Stem Cells and the Differentiation Hierarchy in Mammary Gland Development. Physiol Rev 2019; 100:489-523. [PMID: 31539305 DOI: 10.1152/physrev.00040.2018] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The mammary gland is a highly dynamic organ that undergoes profound changes within its epithelium during puberty and the reproductive cycle. These changes are fueled by dedicated stem and progenitor cells. Both short- and long-lived lineage-restricted progenitors have been identified in adult tissue as well as a small pool of multipotent mammary stem cells (MaSCs), reflecting intrinsic complexity within the epithelial hierarchy. While unipotent progenitor cells predominantly execute day-to-day homeostasis and postnatal morphogenesis during puberty and pregnancy, multipotent MaSCs have been implicated in coordinating alveologenesis and long-term ductal maintenance. Nonetheless, the multipotency of stem cells in the adult remains controversial. The advent of large-scale single-cell molecular profiling has revealed striking changes in the gene expression landscape through ontogeny and the presence of transient intermediate populations. An increasing number of lineage cell-fate determination factors and potential niche regulators have now been mapped along the hierarchy, with many implicated in breast carcinogenesis. The emerging diversity among stem and progenitor populations of the mammary epithelium is likely to underpin the heterogeneity that characterizes breast cancer.
Collapse
Affiliation(s)
- Nai Yang Fu
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Emma Nolan
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Geoffrey J Lindeman
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Jane E Visvader
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
17
|
Affiliation(s)
- Philip Bland
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Beatrice A Howard
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK.
| |
Collapse
|
18
|
Rozenberg JM, Taylor JM, Mack CP. RBPJ binds to consensus and methylated cis elements within phased nucleosomes and controls gene expression in human aortic smooth muscle cells in cooperation with SRF. Nucleic Acids Res 2019; 46:8232-8244. [PMID: 29931229 PMCID: PMC6144787 DOI: 10.1093/nar/gky562] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 06/07/2018] [Indexed: 11/15/2022] Open
Abstract
Given our previous demonstration that RBPJ binds a methylated repressor element and regulates smooth muscle cell (SMC)-specific gene expression, we used genome-wide approaches to identify RBPJ binding regions in human aortic SMC and to assess RBPJ's effects on chromatin structure and gene expression. RBPJ bound to consensus cis elements, but also to TCmGGGA sequences within Alu repeats that were less transcriptionally active as assessed by DNAse hypersensitivity, H3K9 acetylation, and Notch3 and RNA Pol II binding. Interestingly, RBPJ binding was frequently detected at the borders of open chromatin, and a large fraction of genes induced or repressed by RBPJ depletion were associated with this cluster of RBPJ binding sites. RBPJ binding dramatically co-localized with serum response factor (SRF) and RNA seq experiments in RBPJ- and SRF-depleted SMC demonstrated that these factors interact functionally to regulate the contraction and inflammatory gene programs that help define SMC phenotype. Finally, we showed that RBPJ bound preferentially to phased nucleosomes independent of active chromatin marks and to cis elements positioned at the beginning and middle of the nucleosome dyad. These novel findings add important insight into RBPJ's role in chromatin structure and gene expression in SMC.
Collapse
Affiliation(s)
- Julian M Rozenberg
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Joan M Taylor
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Christopher P Mack
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
19
|
Slepicka PF, Cyrill SL, Dos Santos CO. Pregnancy and Breast Cancer: Pathways to Understand Risk and Prevention. Trends Mol Med 2019; 25:866-881. [PMID: 31383623 DOI: 10.1016/j.molmed.2019.06.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/10/2019] [Accepted: 06/17/2019] [Indexed: 12/14/2022]
Abstract
Several studies have made strong efforts to understand how age and parity modulate the risk of breast cancer. A holistic understanding of the dynamic regulation of the morphological, cellular, and molecular milieu of the mammary gland offers insights into the drivers of breast cancer development as well as into potential prophylactic interventions, the latter being a longstanding ambition of the research and clinical community aspiring to eradicate the disease. In this review we discuss mechanisms that react to pregnancy signals, and we delineate the nuances of pregnancy-associated dynamism that contribute towards either breast cancer development or prevention. Further definition of the molecular basis of parity and breast cancer risk may allow the elaboration of tools to predict and survey those who are at risk of breast cancer development.
Collapse
Affiliation(s)
- Priscila F Slepicka
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Samantha L Cyrill
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Camila O Dos Santos
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
20
|
Downregulation of Notch Signaling in Kras-Induced Gastric Metaplasia. Neoplasia 2019; 21:810-821. [PMID: 31276933 PMCID: PMC6611983 DOI: 10.1016/j.neo.2019.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/28/2019] [Accepted: 06/04/2019] [Indexed: 11/21/2022] Open
Abstract
Activating mutations and amplification of Kras and, more frequently, signatures for Kras activation are noted in stomach cancer. Expression of mutant KrasG12D in the mouse gastric mucosa has been shown to induce hyperplasia and metaplasia. However, the mechanisms by which Kras activation leads to gastric metaplasia are not fully understood. Here we report that KrasLSL-G12D/+;Pdx1-cre, a mouse model known for pancreatic cancer, also mediates KrasG12D expression in the stomach, causing gastric hyperplasia and metaplasia prior to the pathologic changes in the pancreas. These mice exhibit ectopic cell proliferation at the base of gastric glands, whereas wild-type mice contain proliferating cells primarily at the isthmus/neck of the gastric glands. Notch signaling is decreased in the KrasLSL-G12D/+;Pdx1-cre gastric mucosa, as shown by lower levels of cleaved Notch intracellular domains and downregulation of Notch downstream target genes. Expression of a Notch ligand Jagged1 is downregulated at the base of the mutant gland, accompanied by loss of chief cell marker Mist1. We demonstrate that exogenous Jagged1 or overexpression of Notch intracellular domain stimulates Mist1 expression in gastric cancer cell lines, suggesting positive regulation of Mist1 by Notch signaling. Finally, deletion of Jagged1 or Notch3 in KrasLSL-G12D/+;Pdx1-cre mice promoted development of squamous cell carcinoma in the forestomach, albeit short of invasive adenocarcinoma in the glandular stomach. Taken together, these results reveal downregulation of Notch signaling and Mist1 expression during the initiation of Kras-driven gastric tumorigenesis and suggest a tumor-suppressive role for Notch in this context.
Collapse
|
21
|
Spatially correlated phenotyping reveals K5-positive luminal progenitor cells and p63-K5/14-positive stem cell-like cells in human breast epithelium. J Transl Med 2018; 98:1065-1075. [PMID: 29743728 DOI: 10.1038/s41374-018-0054-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 01/19/2018] [Accepted: 02/18/2018] [Indexed: 01/15/2023] Open
Abstract
Understanding the mechanisms regulating human mammary epithelium requires knowledge of the cellular constituents of this tissue. Different and partially contradictory definitions and concepts describing the cellular hierarchy of mammary epithelium have been proposed, including our studies of keratins K5 and/or K14 as markers of progenitor cells. Furthermore, we and others have suggested that the p53 homolog p63 is a marker of human breast epithelial stem cells. In this investigation, we expand our previous studies by testing whether immunohistochemical staining with monospecific anti-keratin antibodies in combination with an antibody against the stem cell marker p63 might help refine the different morphologic phenotypes in normal breast epithelium. We used in situ multilabel staining for p63, different keratins, the myoepithelial marker smooth muscle actin (SMA), the estrogen receptor (ER), and Ki67 to dissect and quantify the cellular components of 16 normal pre- and postmenopausal human breast epithelial tissue samples at the single-cell level. Importantly, we confirm the existence of K5+ only cells and suggest that they, in contrast to the current view, are key luminal precursor cells from which K8/18+ progeny cells evolve. These cells are further modified by the expression of ER and Ki67. We have also identified a population of p63+K5+ cells that are only found in nipple ducts. Based on our findings, we propose a new concept of the cellular hierarchy of human breast epithelium, including K5 luminal lineage progenitors throughout the ductal-lobular axis and p63+K5+ progenitors confined to the nipple ducts.
Collapse
|
22
|
Suen AA, Jefferson WN, Williams CJ, Wood CE. Differentiation Patterns of Uterine Carcinomas and Precursor Lesions Induced by Neonatal Estrogen Exposure in Mice. Toxicol Pathol 2018; 46:574-596. [PMID: 29895210 PMCID: PMC6027618 DOI: 10.1177/0192623318779326] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Developmental exposure to estrogenic chemicals is an established risk factor for cancer of the female reproductive tract. This increase in risk has been associated with disruption of normal patterns of cellular differentiation during critical stages of morphogenesis. The goal of this study was to document uterine epithelial phenotypes over time following neonatal treatment with the synthetic estrogen diethylstilbestrol (DES) or the soy phytoestrogen genistein (GEN) in female CD-1 mice. Both DES and GEN induced three distinct populations of abnormal endometrial epithelial cells: luminal (SIX1+/P63-/CK14-/CK18+), basal (SIX1+/P63+/CK14+/CK18-), and mixed/bipotential (SIX1+/P63-/CK14+/CK18+), which were all established by early adulthood. In older animals, DES and GEN resulted in uterine carcinomas with mixed glandular, basal, and squamous cell elements. All carcinomas were composed largely of the three abnormal cell types. These findings identify novel epithelial differentiation patterns in the uterus and support the idea that disruption of cellular programming in early development can influence cancer risk later in life.
Collapse
Affiliation(s)
- Alisa A. Suen
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
- Oak Ridge Institute for Science and Education (ORISE) participant in the Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Wendy N. Jefferson
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Carmen J. Williams
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Charles E. Wood
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| |
Collapse
|
23
|
Notch inhibitors and their role in the treatment of triple negative breast cancer: promises and failures. Curr Opin Oncol 2017; 29:411-427. [DOI: 10.1097/cco.0000000000000406] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
24
|
Abstract
The mammary epithelium is organized in a hierarchy of mammary stem cells (MaSCs), progenitors, and differentiated cells. The development and homeostasis of mammary gland are tightly controlled by a complex network of cell lineage regulators. These determinants of cellular hierarchy are frequently deregulated in breast tumor cells and closely associated with cancer progression and metastasis. They also contribute to the diversity of breast cancer subtypes and their distinct metastatic patterns. Cell fate regulators that normally promote stem/progenitor activities can serve as drivers for epithelial-mesenchymal transition and metastasis whereas regulators that promote terminal differentiation generally suppress metastasis. In this review, we discuss how some of the key factors function in normal mammary lineage determination and how these processes are hijacked by tumor cells to enhance metastasis. Understanding the molecular connections between normal development and cancer metastasis will enable the development of more specific and effective therapeutic approaches targeting metastatic tumor cells.
Collapse
Affiliation(s)
- Wei Lu
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ, 08544, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ, 08544, USA.
| |
Collapse
|
25
|
Multicolor immunofluorescence reveals that p63- and/or K5-positive progenitor cells contribute to normal breast epithelium and usual ductal hyperplasia but not to low-grade intraepithelial neoplasia of the breast. Virchows Arch 2017; 470:493-504. [PMID: 28303349 DOI: 10.1007/s00428-017-2073-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 12/01/2016] [Accepted: 01/16/2017] [Indexed: 10/20/2022]
Abstract
We contend that knowledge about the cellular composition of normal breast epithelium is a prerequisite for understanding proliferative breast disease. Against this background, we used multicolor immunofluorescence to study normal breast epithelium and two types of intraepithelial proliferative breast lesion for expression of the p63, basal keratin K5, glandular keratin K8/18, SMA, ER-alpha, and Ki67. We studied eight normal breast epithelium samples, 12 cases of usual ductal hyperplasia, and 33 cases of low-grade intraepithelial neoplasia (9 flat epithelial atypia, 14 low-grade ductal carcinoma in situ and 10 cases of lobular neoplasia). Usual ductal hyperplasia showed striking similarity to normal luminal breast epithelium including p63+ and/or K5+ luminal progenitor cells and the full spectrum of luminal progeny cells. In normal breast epithelium and usual ductal hyperplasia, expression of ER-alpha was associated with lack of expression of the proliferation antigen Ki67. In contrast, we found in both types of low-grade intraepithelial neoplasia robust expression of keratin K8/18 and a positive association between ER-alpha and Ki67 expression. However, these lesions were consistently negative for p63 and/or K5. Our observational study supports the view that usual ductal hyperplasia and low-grade intraepithelial neoplasia are different entities rather than part of a spectrum of the same disease. We propose a new operational model of cell differentiation that may serve to better understand correlations between normal breast epithelium and proliferative breast diseases. From our data we conclude that p63+ and/or K5+ progenitor cells contribute to maintenance of normal epithelium and usual ductal hyperplasia, but not to low-grade intraepithelial neoplasia of the breast.
Collapse
|
26
|
Shi FT, Yu M, Zloty D, Bell RH, Wang E, Akhoundsadegh N, Leung G, Haegert A, Carr N, Shapiro J, McElwee KJ. Notch signaling is significantly suppressed in basal cell carcinomas and activation induces basal cell carcinoma cell apoptosis. Mol Med Rep 2017; 15:1441-1454. [PMID: 28259916 PMCID: PMC5364965 DOI: 10.3892/mmr.2017.6163] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 09/09/2016] [Indexed: 12/29/2022] Open
Abstract
A subset of basal cell carcinomas (BCCs) are directly derived from hair follicles (HFs). In some respects, HFs can be defined as 'ordered' skin appendage growths, while BCCs can be regarded as 'disordered' skin appendage growths. The aim of the present study was to examine HFs and BCCs to define the expression of common and unique signaling pathways in each skin appendage. Human nodular BCCs, along with HFs and non‑follicular skin epithelium from normal individuals, were examined using microarrays, qPCR, and immunohistochemistry. Subsequently, BCC cells and root sheath keratinocyte cells from HFs were cultured and treated with Notch signaling peptide Jagged1 (JAG1). Gene expression, protein levels, and cell apoptosis susceptibility were assessed using qPCR, immunoblotting, and flow cytometry, respectively. Specific molecular mechanisms were found to be involved in the process of cell self‑renewal in the HFs and BCCs, including Notch and Hedgehog signaling pathways. However, several key Notch signaling factors showed significant differential expression in BCCs compared with HFs. Stimulating Notch signaling with JAG1 induced apoptosis of BCC cells by increasing Fas ligand expression and downstream caspase-8 activation. The present study showed that Notch signaling pathway activity is suppressed in BCCs, and is highly expressed in HFs. Elements of the Notch pathway could, therefore, represent targets for the treatment of BCCs and potentially in hair follicle engineering.
Collapse
Affiliation(s)
- Feng-Tao Shi
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V5Z 4E8, Canada
| | - Mei Yu
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V5Z 4E8, Canada
| | - David Zloty
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V5Z 4E8, Canada
| | - Robert H Bell
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC V6H 3Z6, Canada
| | - Eddy Wang
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V5Z 4E8, Canada
| | - Noushin Akhoundsadegh
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V5Z 4E8, Canada
| | - Gigi Leung
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V5Z 4E8, Canada
| | - Anne Haegert
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC V6H 3Z6, Canada
| | - Nicholas Carr
- Department of Surgery, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Jerry Shapiro
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V5Z 4E8, Canada
| | - Kevin J McElwee
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V5Z 4E8, Canada
| |
Collapse
|
27
|
Fundamental Pathways in Breast Cancer 4: Signaling to Chromatin in Breast Development. Breast Cancer 2017. [DOI: 10.1007/978-3-319-48848-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
28
|
Dou XW, Liang YK, Lin HY, Wei XL, Zhang YQ, Bai JW, Chen CF, Chen M, Du CW, Li YC, Tian J, Man K, Zhang GJ. Notch3 Maintains Luminal Phenotype and Suppresses Tumorigenesis and Metastasis of Breast Cancer via Trans-Activating Estrogen Receptor-α. Theranostics 2017; 7:4041-4056. [PMID: 29109797 PMCID: PMC5667424 DOI: 10.7150/thno.19989] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 08/11/2017] [Indexed: 02/05/2023] Open
Abstract
The luminal A phenotype is the most common breast cancer subtype and is characterized by estrogen receptor α expression (ERα). Identification of the key regulator that governs the luminal phenotype of breast cancer will clarify the pathogenic mechanism and provide novel therapeutic strategies for this subtype of cancer. ERα signaling pathway sustains the epithelial phenotype and inhibits the epithelial-mesenchymal transition (EMT) of breast cancer. In this study, we demonstrate that Notch3 positively associates with ERα in both breast cancer cell lines and human breast cancer tissues. We found that overexpression of Notch3 intra-cellular domain, a Notch3 active form (N3ICD), in ERα negative breast cancer cells re-activated ERα, while knock-down of Notch3 reduced ERα transcript and proteins, with alteration of down-stream genes, suggesting its ability to regulate ERα. Mechanistically, our results show that Notch3 specifically binds to the CSL binding element of the ERα promoter and activates ERα expression. Moreover, Notch3 suppressed EMT, while suppression of Notch3 promoted EMT in cellular assay. Overexpressing N3ICD in triple-negative breast cancer suppressed tumorigenesis and metastasis in vivo. Conversely, depletion of Notch3 in luminal breast cancer promoted metastasis in vivo. Furthermore, Notch3 transcripts were significantly associated with prolonged relapse-free survival in breast cancer, in particular in ERα positive breast cancer patients. Our observations demonstrate that Notch3 governs the luminal phenotype via trans-activating ERα expression in breast cancer. These findings delineate the role of a Notch3/ERα axis in maintaining the luminal phenotype and inhibiting tumorigenesis and metastasis in breast cancer, providing a novel strategy to re-sensitize ERα negative or low-expressing breast cancers to hormone therapy.
Collapse
Affiliation(s)
- Xiao-Wei Dou
- The Breast Center, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
| | - Yuan-Ke Liang
- The Breast Center, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Hao-Yu Lin
- The Breast Center, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
- Department of Breast and Thyroid Surgery, the First Affiliated Hospital of SUMC
| | - Xiao-Long Wei
- The Breast Center, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
- Department of Pathology, the Cancer Hospital of Shantou University Medical College (SUMC), China
| | - Yong-Qu Zhang
- The Breast Center, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
| | - Jing-Wen Bai
- The Breast Center, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
| | - Chun-Fa Chen
- The Breast Center, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
| | - Min Chen
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
| | - Cai-Wen Du
- Department of Breast Medical Oncology, the Cancer Hospital of Shantou University Medical College (SUMC), China
| | - Yao-Chen Li
- The Breast Center, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
| | - Jie Tian
- Institute of Automation, Chinese Academy of Science, China
| | - Kwan Man
- Department of Surgery, Hong Kong University Li Ka-Tsing faculty of Medicine, Hong Kong, China
| | - Guo-Jun Zhang
- The Breast Center, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ✉ Corresponding author: Guo-Jun Zhang, MD, PhD. Tel.: +86(754)88556826; E-mail:
| |
Collapse
|
29
|
Santoro A, Vlachou T, Carminati M, Pelicci PG, Mapelli M. Molecular mechanisms of asymmetric divisions in mammary stem cells. EMBO Rep 2016; 17:1700-1720. [PMID: 27872203 DOI: 10.15252/embr.201643021] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/04/2016] [Accepted: 10/25/2016] [Indexed: 01/16/2023] Open
Abstract
Stem cells have the remarkable ability to undergo proliferative symmetric divisions and self-renewing asymmetric divisions. Balancing of the two modes of division sustains tissue morphogenesis and homeostasis. Asymmetric divisions of Drosophila neuroblasts (NBs) and sensory organ precursor (SOP) cells served as prototypes to learn what we consider now principles of asymmetric mitoses. They also provide initial evidence supporting the notion that aberrant symmetric divisions of stem cells could correlate with malignancy. However, transferring the molecular knowledge of circuits underlying asymmetry from flies to mammals has proven more challenging than expected. Several experimental approaches have been used to define asymmetry in mammalian systems, based on daughter cell fate, unequal partitioning of determinants and niche contacts, or proliferative potential. In this review, we aim to provide a critical evaluation of the assays used to establish the stem cell mode of division, with a particular focus on the mammary gland system. In this context, we will discuss the genetic alterations that impinge on the modality of stem cell division and their role in breast cancer development.
Collapse
Affiliation(s)
- Angela Santoro
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Thalia Vlachou
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Manuel Carminati
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | | | - Marina Mapelli
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| |
Collapse
|
30
|
Callahan R, Chestnut BA, Raafat A. Original Research: Featured Article: Imatinib mesylate (Gleevec) inhibits Notch and c-Myc signaling: Five-day treatment permanently rescues mammary development. Exp Biol Med (Maywood) 2016; 242:53-67. [PMID: 27550925 DOI: 10.1177/1535370216665175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/28/2016] [Indexed: 12/31/2022] Open
Abstract
Wap-Int3 transgenic females expressing the Notch4 intracellular domain (designated Int3) from the whey acidic protein promoter exhibit two phenotypes in the mammary gland: blockage of lobuloalveolar development and lactation, and tumor development with 100% penetrance. Previously, we have shown that treatment of Wap-Int3 tumor bearing mice with Imatinib mesylate (Gleevec) is associated with complete regression of the tumor. In the present study, we show that treatment of Wap-Int3 mice during day 1 through day 6 of pregnancy with Gleevec leads to the restoration of their lobuloalveolar development and ability to lactate in subsequent pregnancies in absence of Gleevec treatment. In addition, these mice do not develop mammary tumors. We investigated the mechanism for Gleevec regulation of Notch signaling and found that Gleevec treatment results in a loss of Int3 protein but not of Int3 mRNA in HC11 mouse mammary epithelial cells expressing Int3. The addition of MG-132, a proteasome inhibitor, shows increased ubiquitination of Int3 in the presence of Gleevec. Thus, Gleevec affects the stability of Int3 by promoting the degradation of Int3 via E3 ubiquitin ligases targeting it for the proteasome degradation. Gleevec is a tyrosine kinase inhibitor that acts on c-Kit and PDGFR. Therefore, we investigated the downstream substrate kinase GSK3β to ascertain the possible role that this kinase might play in the stability of Int3. Data show that Gleevec degradation of Int3 is GSK3β dependent. We have expanded our study of the effects Gleevec has on tumorigenesis of other oncogenes. We have found that anchorage-independent growth of HC11-c-Myc cells as well as tumor growth in nude mice is inhibited by Gleevec treatment. As with Int3, Gleevec treatment appears to destabilize the c-Myc protein but not mRNA. These results indicate that Gleevec could be a potential therapeutic drug for patients bearing Notch4 and/or c-Myc positive breast carcinomas.
Collapse
Affiliation(s)
- Robert Callahan
- Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Barry A Chestnut
- Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Ahmed Raafat
- Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| |
Collapse
|
31
|
Histone Demethylase KDM6A Controls the Mammary Luminal Lineage through Enzyme-Independent Mechanisms. Mol Cell Biol 2016; 36:2108-20. [PMID: 27215382 DOI: 10.1128/mcb.00089-16] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/08/2016] [Indexed: 02/06/2023] Open
Abstract
Establishment of the mammary luminal cell lineage is controlled primarily by hormones and through specific transcription factors (TFs). Previous studies have linked histone methyltransferases to the differentiation of mammary epithelium, thus opening the possibility of biological significance of counteracting demethylases. We have now demonstrated an essential role for the H3K27me3 demethylase KDM6A in generating a balanced alveolar compartment. Deletion of Kdm6a in the mammary luminal cell lineage led to a paucity of luminal cells and an excessive expansion of basal cells, both in vivo and in vitro The inability to form structurally normal ducts and alveoli during pregnancy resulted in lactation failure. Mutant luminal cells did not exhibit their distinctive transcription factor pattern and displayed basal characteristics. The genomic H3K27me3 landscape was unaltered in mutant tissue, and support for a demethylase-independent mechanism came from mice expressing a catalytically inactive KDM6A. Mammary tissue developed normally in these mice. Chromatin immunoprecipitation sequencing (ChIP-seq) experiments demonstrated KDM6A binding to putative enhancers enriched for key mammary TFs and H3K27ac. This study demonstrated for the first time that the mammary luminal lineage relies on KDM6A to ensure a transcription program leading to differentiated alveoli. Failure to fully implement this program results in structurally and functionally impaired mammary tissue.
Collapse
|
32
|
Cantrell MA, Ebelt ND, Pfefferle AD, Perou CM, Van Den Berg CL. c-Jun N-terminal kinase 2 prevents luminal cell commitment in normal mammary glands and tumors by inhibiting p53/Notch1 and breast cancer gene 1 expression. Oncotarget 2016; 6:11863-81. [PMID: 25970777 PMCID: PMC4494910 DOI: 10.18632/oncotarget.3787] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 04/13/2015] [Indexed: 12/19/2022] Open
Abstract
Breast cancer is a heterogeneous disease with several subtypes carrying unique prognoses. Patients with differentiated luminal tumors experience better outcomes, while effective treatments are unavailable for poorly differentiated tumors, including the basal-like subtype. Mechanisms governing mammary tumor subtype generation could prove critical to developing better treatments. C-Jun N-terminal kinase 2 (JNK2) is important in mammary tumorigenesis and tumor progression. Using a variety of mouse models, human breast cancer cell lines and tumor expression data, studies herein support that JNK2 inhibits cell differentiation in normal and cancer-derived mammary cells. JNK2 prevents precocious pubertal mammary development and inhibits Notch-dependent expansion of luminal cell populations. Likewise, JNK2 suppresses luminal populations in a p53-competent Polyoma Middle T-antigen tumor model where jnk2 knockout causes p53-dependent upregulation of Notch1 transcription. In a p53 knockout model, JNK2 restricts luminal populations independently of Notch1, by suppressing Brca1 expression and promoting epithelial to mesenchymal transition. JNK2 also inhibits estrogen receptor (ER) expression and confers resistance to fulvestrant, an ER inhibitor, while stimulating tumor progression. These data suggest that therapies inhibiting JNK2 in breast cancer may promote tumor differentiation, improve endocrine therapy response, and inhibit metastasis.
Collapse
Affiliation(s)
- Michael A Cantrell
- Institute of Cellular & Molecular Biology, College of Pharmacy, University of Texas at Austin, Dell Pediatric Research Institute, Austin, TX 78723, USA
| | - Nancy D Ebelt
- Institute of Cellular & Molecular Biology, College of Pharmacy, University of Texas at Austin, Dell Pediatric Research Institute, Austin, TX 78723, USA
| | - Adam D Pfefferle
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC 27599, USA
| | - Charles M Perou
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC 27599, USA.,Department of Genetics, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC 27599, USA
| | - Carla Lynn Van Den Berg
- Institute of Cellular & Molecular Biology, College of Pharmacy, University of Texas at Austin, Dell Pediatric Research Institute, Austin, TX 78723, USA.,Division of Pharmacology &Toxicology, College of Pharmacy, University of Texas at Austin, Dell Pediatric Research Institute, Austin, TX 78723, USA
| |
Collapse
|
33
|
Bhat V, Sun YJ, Weger S, Raouf A. Notch-Induced Expression of FZD7 Requires Noncanonical NOTCH3 Signaling in Human Breast Epithelial Cells. Stem Cells Dev 2016; 25:522-9. [PMID: 26847503 DOI: 10.1089/scd.2015.0315] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The evolutionarily conserved Notch and Wnt signaling pathways have demonstrated roles in normal mammary gland development and in breast carcinogenesis. We previously reported that in human mammary gland, signaling through NOTCH3 alone regulates the commitment of the undifferentiated bipotential progenitors to the luminal cell fate, indicating that NOTCH3 may regulate the expression of unique genes apart from the other Notch receptors. In this study, we used gain of function and loss of function experiments and found that a Wnt signaling receptor, Frizzled7 (FZD7), is a unique and nonredundant target of NOTCH3 in human breast epithelial cells. Interestingly, neither the constitutively active forms of NOTCH1-2, 4 nor loss of expression of these receptors were able to alter expression of FZD7 in human breast epithelial cells. We further show that FZD7-expressing cells are found more frequently in the luminal progenitor-enriched subpopulation of cells obtained from breast reduction samples compared with the undifferentiated bipotent progenitors. Also, we show that NOTCH3-induced expression of FZD7 occurs in the absence of CSL (CBF1-Suppressor of Hairless-Lag-1). Our data suggest that noncanonical Notch signaling through NOTCH3 could modulate Wnt signaling via FZD7 and in this way, might be involved in luminal cell differentiation.
Collapse
Affiliation(s)
- Vasudeva Bhat
- 1 Department of Immunology, Faculty of Health Sciences, University of Manitoba , Winnipeg, Manitoba, Canada .,2 Research Institute for Oncology and Hematology, CancerCare Manitoba , Winnipeg, Manitoba, Canada
| | - Yu Jia Sun
- 1 Department of Immunology, Faculty of Health Sciences, University of Manitoba , Winnipeg, Manitoba, Canada .,2 Research Institute for Oncology and Hematology, CancerCare Manitoba , Winnipeg, Manitoba, Canada
| | - Steve Weger
- 1 Department of Immunology, Faculty of Health Sciences, University of Manitoba , Winnipeg, Manitoba, Canada .,2 Research Institute for Oncology and Hematology, CancerCare Manitoba , Winnipeg, Manitoba, Canada
| | - Afshin Raouf
- 1 Department of Immunology, Faculty of Health Sciences, University of Manitoba , Winnipeg, Manitoba, Canada .,2 Research Institute for Oncology and Hematology, CancerCare Manitoba , Winnipeg, Manitoba, Canada
| |
Collapse
|
34
|
Rangel MC, Bertolette D, Castro NP, Klauzinska M, Cuttitta F, Salomon DS. Developmental signaling pathways regulating mammary stem cells and contributing to the etiology of triple-negative breast cancer. Breast Cancer Res Treat 2016; 156:211-26. [PMID: 26968398 PMCID: PMC4819564 DOI: 10.1007/s10549-016-3746-7] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 03/04/2016] [Indexed: 12/17/2022]
Abstract
Cancer has been considered as temporal and spatial aberrations of normal development in tissues. Similarities between mammary embryonic development and cell transformation suggest that the underlying processes required for mammary gland development are also those perturbed during various stages of mammary tumorigenesis and breast cancer (BC) development. The master regulators of embryonic development Cripto-1, Notch/CSL, and Wnt/β-catenin play key roles in modulating mammary gland morphogenesis and cell fate specification in the embryo through fetal mammary stem cells (fMaSC) and in the adult organism particularly within the adult mammary stem cells (aMaSC), which determine mammary progenitor cell lineages that generate the basal/myoepithelial and luminal compartments of the adult mammary gland. Together with recognized transcription factors and embryonic stem cell markers, these embryonic regulatory molecules can be inappropriately augmented during tumorigenesis to support the tumor-initiating cell (TIC)/cancer stem cell (CSC) compartment, and the effects of their deregulation may contribute for the etiology of BC, in particular the most aggressive subtype of BC, triple-negative breast cancer (TNBC). This in depth review will present evidence of the involvement of Cripto-1, Notch/CSL, and Wnt/β-catenin in the normal mammary gland morphogenesis and tumorigenesis, from fMaSC/aMaSC regulation to TIC generation and maintenance in TNBC. Specific therapies for treating TNBC by targeting these embryonic pathways in TICs will be further discussed, providing new opportunities to destroy not only the bulk tumor, but also TICs that initiate and promote the metastatic spread and recurrence of this aggressive subtype of BC.
Collapse
Affiliation(s)
- Maria Cristina Rangel
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-40B, 1050 Boyles Street, Ft. Detrick, Frederick, MD, 21702, USA
| | - Daniel Bertolette
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-40B, 1050 Boyles Street, Ft. Detrick, Frederick, MD, 21702, USA
| | - Nadia P Castro
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-40B, 1050 Boyles Street, Ft. Detrick, Frederick, MD, 21702, USA
| | - Malgorzata Klauzinska
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-40B, 1050 Boyles Street, Ft. Detrick, Frederick, MD, 21702, USA
| | - Frank Cuttitta
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-40B, 1050 Boyles Street, Ft. Detrick, Frederick, MD, 21702, USA
| | - David S Salomon
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-40B, 1050 Boyles Street, Ft. Detrick, Frederick, MD, 21702, USA.
| |
Collapse
|
35
|
Abstract
Many cancers have similar aberrations in various signaling cascades with crucial roles in cellular proliferation, differentiation, and morphogenesis. Dysregulation of signal cascades that play integral roles during early cellular development is well known to be a central feature of many malignancies. One such signaling cascade is the Wnt/β-catenin pathway, which has a profound effect on stem cell proliferation, migration, and differentiation. This pathway is dysregulated in numerous cell types, underscoring its global oncogenetic potential. This review highlights regulators and downstream effectors of this receptor cascade and addresses the increasingly apparent crosstalk of Wnt with other tumorigenic signaling pathways. As understanding of the genetic and epigenetic changes unique to these malignancies increases, identifying the regulatory mechanisms unique to the Wnt/β-catenin pathway and similarly aberrant receptor pathways will be imperative.
Collapse
Affiliation(s)
- Saint-Aaron L Morris
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Suyun Huang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA; Program in Cancer Biology, The University of Texas Graduate School of Biomedical Sciences, 6767 Bertner Avenue, Mitchell Building BSRB S3.8344, Houston, TX 77030, USA
| |
Collapse
|
36
|
Li XY, Zhai WJ, Teng CB. Notch Signaling in Pancreatic Development. Int J Mol Sci 2015; 17:ijms17010048. [PMID: 26729103 PMCID: PMC4730293 DOI: 10.3390/ijms17010048] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 12/22/2015] [Accepted: 12/24/2015] [Indexed: 12/12/2022] Open
Abstract
The Notch signaling pathway plays a significant role in embryonic cell fate determination and adult tissue homeostasis. Various studies have demonstrated the deep involvement of Notch signaling in the development of the pancreas and the lateral inhibition of Notch signaling in pancreatic progenitor differentiation and maintenance. The targeted inactivation of the Notch pathway components promotes premature differentiation of the endocrine pancreas. However, there is still the contrary opinion that Notch signaling specifies the endocrine lineage. Here, we review the current knowledge of the Notch signaling pathway in pancreatic development and its crosstalk with the Wingless and INT-1 (Wnt) and fibroblast growth factor (FGF) pathways.
Collapse
Affiliation(s)
- Xu-Yan Li
- College of Life Science, Northeast Forestry University, Harbin 150040, China.
- College of Life Sciences, Agriculture and Forestry, Qiqihar University, Qiqihar 161006, China.
| | - Wen-Jun Zhai
- College of Life Science, Northeast Forestry University, Harbin 150040, China.
| | - Chun-Bo Teng
- College of Life Science, Northeast Forestry University, Harbin 150040, China.
| |
Collapse
|
37
|
Hilton HN, Graham JD, Clarke CL. Minireview: Progesterone Regulation of Proliferation in the Normal Human Breast and in Breast Cancer: A Tale of Two Scenarios? Mol Endocrinol 2015; 29:1230-42. [PMID: 26266959 DOI: 10.1210/me.2015-1152] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Progesterone (P), which signals through the P receptor (PR), is critical in normal development of the breast, but its signaling axis is also a major driver of breast cancer risk. Here we review recent advances in the understanding of P signaling in the normal human breast, with a focus on the importance of the balance between autocrine and paracrine signaling. To date, most data (which derive largely from mouse models or human breast cancer cell line studies) have demonstrated that the vast majority of PR+ cells appear to act as "sensor" cells, which respond to P stimulation by translating these hormonal cues into paracrine signals. However, growing evidence suggests that, dependent on the cellular context, P may also signal in an autocrine manner in a subset of cells in the normal mouse mammary gland and human breast. It has been suggested that it may be dysregulation of this autocrine signaling, resulting in a "switch" from a predominance of paracrine signaling to autocrine signaling in PR+ cells, which is an early event during breast tumorigenesis. This review summarizes current evidence in the literature that demonstrates the mechanisms through which P acts in the normal human breast, as well as highlighting the important questions that remain unanswered.
Collapse
Affiliation(s)
- Heidi N Hilton
- Westmead Millennium Institute, University of Sydney, Westmead, New South Wales, 2145, Australia
| | - J Dinny Graham
- Westmead Millennium Institute, University of Sydney, Westmead, New South Wales, 2145, Australia
| | - Christine L Clarke
- Westmead Millennium Institute, University of Sydney, Westmead, New South Wales, 2145, Australia
| |
Collapse
|
38
|
Wang K, Zhang Q, Li D, Ching K, Zhang C, Zheng X, Ozeck M, Shi S, Li X, Wang H, Rejto P, Christensen J, Olson P. PEST domain mutations in Notch receptors comprise an oncogenic driver segment in triple-negative breast cancer sensitive to a γ-secretase inhibitor. Clin Cancer Res 2015; 21:1487-96. [PMID: 25564152 DOI: 10.1158/1078-0432.ccr-14-1348] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To identify and characterize novel, activating mutations in Notch receptors in breast cancer and to determine response to the gamma secretase inhibitor (GSI) PF-03084014. EXPERIMENTAL DESIGN We used several computational approaches, including novel algorithms, to analyze next-generation sequencing data and related omic datasets from The Cancer Genome Atlas (TCGA) breast cancer cohort. Patient-derived xenograft (PDX) models were sequenced, and Notch-mutant models were treated with PF-03084014. Gene-expression and functional analyses were performed to study the mechanism of activation through mutation and inhibition by PF-03084014. RESULTS We identified mutations within and upstream of the PEST domains of NOTCH1, NOTCH2, and NOTCH3 in the TCGA dataset. Mutations occurred via several genetic mechanisms and compromised the function of the PEST domain, a negative regulatory domain commonly mutated in other cancers. Focal amplifications of NOTCH2 and NOTCH3 were also observed, as were heterodimerization or extracellular domain mutations at lower incidence. Mutations and amplifications often activated the Notch pathway as evidenced by increased expression of canonical Notch target genes, and functional mutations were significantly enriched in the triple-negative breast cancer subtype (TNBC). PDX models were also identified that harbored PEST domain mutations, and these models were highly sensitive to PF-03084014. CONCLUSIONS This work suggests that Notch-altered breast cancer constitutes a bona fide oncogenic driver segment with the most common alteration being PEST domain mutations present in multiple Notch receptors. Importantly, functional studies suggest that this newly identified class can be targeted with Notch inhibitors, including GSIs.
Collapse
Affiliation(s)
- Kai Wang
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Qin Zhang
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Danan Li
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Keith Ching
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Cathy Zhang
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Xianxian Zheng
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Mark Ozeck
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Stephanie Shi
- External Research Solutions, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Xiaorong Li
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Hui Wang
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Paul Rejto
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - James Christensen
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California
| | - Peter Olson
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, San Diego, California.
| |
Collapse
|
39
|
Abstract
Human bladder cancers harbor deletions and point mutations in genes coding for Notch receptors and proteins involved in Notch signaling. This leads to elevated MAPK pathway activation, as direct Notch-mediated transcription of MAPK phosphatase DUSP is lost. These bladder tumors, with impaired Notch signaling, also show basal differentiation.
Collapse
Affiliation(s)
- Keli Xu
- Tumor Cell Biology Program, Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Darius J Bägli
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Sean E Egan
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A1, Canada.
| |
Collapse
|
40
|
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
|
41
|
Abstract
While it has been known for decades that androgen hormones influence normal breast development and breast carcinogenesis, the underlying mechanisms have only been recently elucidated. To date, most studies have focused on androgen action in breast cancer cell lines, yet these studies represent artificial systems that often do not faithfully replicate/recapitulate the cellular, molecular and hormonal environments of breast tumours in vivo. It is critical to have a better understanding of how androgens act in the normal mammary gland as well as in in vivo systems that maintain a relevant tumour microenvironment to gain insights into the role of androgens in the modulation of breast cancer development. This in turn will facilitate application of androgen-modulation therapy in breast cancer. This is particularly relevant as current clinical trials focus on inhibiting androgen action as breast cancer therapy but, depending on the steroid receptor profile of the tumour, certain individuals may be better served by selectively stimulating androgen action. Androgen receptor (AR) protein is primarily expressed by the hormone-sensing compartment of normal breast epithelium, commonly referred to as oestrogen receptor alpha (ERa (ESR1))-positive breast epithelial cells, which also express progesterone receptors (PRs) and prolactin receptors and exert powerful developmental influences on adjacent breast epithelial cells. Recent lineage-tracing studies, particularly those focussed on NOTCH signalling, and genetic analysis of cancer risk in the normal breast highlight how signalling via the hormone-sensing compartment can influence normal breast development and breast cancer susceptibility. This provides an impetus to focus on the relationship between androgens, AR and NOTCH signalling and the crosstalk between ERa and PR signalling in the hormone-sensing component of breast epithelium in order to unravel the mechanisms behind the ability of androgens to modulate breast cancer initiation and growth.
Collapse
Affiliation(s)
- Gerard A Tarulli
- Dame Roma Mitchell Cancer Research Laboratories (DRMCRL)Faculty of Health Sciences, School of Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Lisa M Butler
- Dame Roma Mitchell Cancer Research Laboratories (DRMCRL)Faculty of Health Sciences, School of Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories (DRMCRL)Faculty of Health Sciences, School of Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Theresa E Hickey
- Dame Roma Mitchell Cancer Research Laboratories (DRMCRL)Faculty of Health Sciences, School of Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
42
|
Owens TW, Rogers RL, Best S, Ledger A, Mooney AM, Ferguson A, Shore P, Swarbrick A, Ormandy CJ, Simpson PT, Carroll JS, Visvader J, Naylor MJ. Runx2 is a novel regulator of mammary epithelial cell fate in development and breast cancer. Cancer Res 2014; 74:5277-5286. [PMID: 25056120 DOI: 10.1158/0008-5472.can-14-0053] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Regulators of differentiated cell fate can offer targets for managing cancer development and progression. Here, we identify Runx2 as a new regulator of epithelial cell fate in mammary gland development and breast cancer. Runx2 is expressed in the epithelium of pregnant mice in a strict temporally and hormonally regulated manner. During pregnancy, Runx2 genetic deletion impaired alveolar differentiation in a manner that disrupted alveolar progenitor cell populations. Conversely, exogenous transgenic expression of Runx2 in mammary epithelial cells blocked milk production, suggesting that the decrease in endogenous Runx2 observed late in pregnancy is necessary for full differentiation. In addition, overexpression of Runx2 drove epithelial-to-mesenchymal transition-like changes in normal mammary epithelial cells, whereas Runx2 deletion in basal breast cancer cells inhibited cellular phenotypes associated with tumorigenesis. Notably, loss of Runx2 expression increased tumor latency and enhanced overall survival in a mouse model of breast cancer, with Runx2-deficient tumors exhibiting reduced cell proliferation. Together, our results establish a previously unreported function for Runx2 in breast cancer that may offer a novel generalized route for therapeutic interventions. Cancer Res; 74(18); 5277-86. ©2014 AACR.
Collapse
Affiliation(s)
- Thomas W Owens
- Discipline of Physiology & Bosch Institute, School of Medical Sciences, The University of Sydney, NSW 2006, Australia
| | - Renee L Rogers
- Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Sarah Best
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria 3052, Australia
| | - Anita Ledger
- Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Anne-Marie Mooney
- Discipline of Physiology & Bosch Institute, School of Medical Sciences, The University of Sydney, NSW 2006, Australia
| | - Alison Ferguson
- Discipline of Physiology & Bosch Institute, School of Medical Sciences, The University of Sydney, NSW 2006, Australia
| | - Paul Shore
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, UK
| | - Alexander Swarbrick
- Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2052, Australia
| | - Christopher J Ormandy
- Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2052, Australia
| | - Peter T Simpson
- The University of Queensland, UQ Centre for Clinical Research (UQCCR), Herston, Queensland 4029, Australia
| | - Jason S Carroll
- Cancer Research UK, Cambridge Research Institute, Cambridge, CB2 0RE, UK
| | - Jane Visvader
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria 3052, Australia
| | - Matthew J Naylor
- Discipline of Physiology & Bosch Institute, School of Medical Sciences, The University of Sydney, NSW 2006, Australia.,Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia.,Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, UK
| |
Collapse
|
43
|
Guo W. Concise review: breast cancer stem cells: regulatory networks, stem cell niches, and disease relevance. Stem Cells Transl Med 2014; 3:942-8. [PMID: 24904174 DOI: 10.5966/sctm.2014-0020] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Accumulating evidence has shown that cancer stem cells (CSCs), the cancer cells that have long-term proliferative potential and the ability to regenerate tumors with phenotypically heterogeneous cell types, are important mediators of tumor metastasis and cancer relapse. In breast cancer, these cells often possess attributes of cells that have undergone an epithelial-mesenchymal transition (EMT). Signaling networks mediated by microRNAs and EMT-inducing transcription factors connect the EMT program with the core stem cell regulatory machineries. These signaling networks are also regulated by extrinsic niche signals that induce and maintain CSCs, contributing to metastatic colonization and promoting the reactivation of dormant tumor cells. Targeting these CSC pathways is likely to improve the efficacy of conventional chemo- and radiotherapies.
Collapse
Affiliation(s)
- Wenjun Guo
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Department of Cell Biology, Albert Einstein College of Medicine, New York, New York, USA
| |
Collapse
|
44
|
Zhang S, Chung WC, Miele L, Xu K. Targeting Met and Notch in the Lfng-deficient, Met-amplified triple-negative breast cancer. Cancer Biol Ther 2014; 15:633-42. [PMID: 24556651 DOI: 10.4161/cbt.28180] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Triple negative breast cancer (TNBC) accounts for 15-20% of breast carcinomas and represents one of the most aggressive forms of this disease. Basal and claudin-low are the two main molecular subtypes among TNBCs. We previously reported that deletion of Lfng in mouse mammary gland caused deregulated Notch activation and induced basal-like and claudin-low tumors with co-selection for Met amplification. In human breast cancers, the vast majority of basal tumors and a subset of claudin-low tumors show reduced Lfng expression. Elevated Met expression and activation is associated with basal as well as claudin-low subtypes. To examine roles of Met and Notch in TNBC cells, we established two cell lines that harbor Met amplification as well as Lfng deletion, and possess features of basal and claudin-low breast cancer subtypes. Pharmacological inhibition of Met not only suppressed cell growth, tumorsphere and colony formation, but also reversed epithelial-to-mesenchymal transition and inhibited cell migration in both cell lines. In contrast, inhibition of Notch signaling using a γ-secretase inhibitor (GSI) only suppressed colony formation. Interestingly, GSI had no effect as single agent, but exerted a synergistic effect with Met inhibitor, on cell growth in 2D culture. We found that inhibition of Met resulted in downregulation of Dll ligands and upregulation of Jagged ligands, leading to differential modulation of Notch signaling. Our results suggest that combination targeting of Met and Notch may prove beneficial for TNBC patients with Met overexpression and Notch hyperactivation.
Collapse
Affiliation(s)
- Shubing Zhang
- Cancer Institute; University of Mississippi Medical Center; Jackson, MS USA
| | - Wen-cheng Chung
- Cancer Institute; University of Mississippi Medical Center; Jackson, MS USA
| | - Lucio Miele
- Cancer Institute; University of Mississippi Medical Center; Jackson, MS USA; Department of Pharmacology and Toxicology; University of Mississippi Medical Center; Jackson, MS USA
| | - Keli Xu
- Cancer Institute; University of Mississippi Medical Center; Jackson, MS USA; Department of Neurobiology and Anatomical Sciences; University of Mississippi Medical Center; Jackson, MS USA
| |
Collapse
|
45
|
Shao Y, Zhao FQ. Emerging evidence of the physiological role of hypoxia in mammary development and lactation. J Anim Sci Biotechnol 2014; 5:9. [PMID: 24444333 PMCID: PMC3929241 DOI: 10.1186/2049-1891-5-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 01/17/2014] [Indexed: 01/22/2023] Open
Abstract
Hypoxia is a physiological or pathological condition of a deficiency of oxygen supply in the body as a whole or within a tissue. During hypoxia, tissues undergo a series of physiological responses to defend themselves against a low oxygen supply, including increased angiogenesis, erythropoiesis, and glucose uptake. The effects of hypoxia are mainly mediated by hypoxia-inducible factor 1 (HIF-1), which is a heterodimeric transcription factor consisting of α and β subunits. HIF-1β is constantly expressed, whereas HIF-1α is degraded under normal oxygen conditions. Hypoxia stabilizes HIF-1α and the HIF complex, and HIF then translocates into the nucleus to initiate the expression of target genes. Hypoxia has been extensively studied for its role in promoting tumor progression, and emerging evidence also indicates that hypoxia may play important roles in physiological processes, including mammary development and lactation. The mammary gland exhibits an increasing metabolic rate from pregnancy to lactation to support mammary growth, lactogenesis, and lactation. This process requires increasing amounts of oxygen consumption and results in localized chronic hypoxia as confirmed by the binding of the hypoxia marker pimonidazole HCl in mouse mammary gland. We hypothesized that this hypoxic condition promotes mammary development and lactation, a hypothesis that is supported by the following several lines of evidence: i) Mice with an HIF-1α deletion selective for the mammary gland have impaired mammary differentiation and lipid secretion, resulting in lactation failure and striking changes in milk compositions; ii) We recently observed that hypoxia significantly induces HIF-1α-dependent glucose uptake and GLUT1 expression in mammary epithelial cells, which may be responsible for the dramatic increases in glucose uptake and GLUT1 expression in the mammary gland during the transition period from late pregnancy to early lactation; and iii) Hypoxia and HIF-1α increase the phosphorylation of signal transducers and activators of transcription 5a (STAT5a) in mammary epithelial cells, whereas STAT5 phosphorylation plays important roles in the regulation of milk protein gene expression and mammary development. Based on these observations, hypoxia effects emerge as a new frontier for studying the regulation of mammary development and lactation.
Collapse
Affiliation(s)
| | - Feng-Qi Zhao
- Laboratory of Lactation and Metabolic Physiology, Department of Animal Science, University of Vermont, Burlington, Vermont 05405, USA.
| |
Collapse
|
46
|
Yallowitz AR, Alexandrova EM, Talos F, Xu S, Marchenko ND, Moll UM. p63 is a prosurvival factor in the adult mammary gland during post-lactational involution, affecting PI-MECs and ErbB2 tumorigenesis. Cell Death Differ 2014; 21:645-54. [PMID: 24440910 DOI: 10.1038/cdd.2013.199] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 12/13/2013] [Accepted: 12/13/2013] [Indexed: 01/31/2023] Open
Abstract
In embryogenesis, p63 is essential to develop mammary glands. In the adult mammary gland, p63 is highly expressed in the basal cell layer that comprises myoepithelial and interspersed stem/progenitor cells, and has limited expression in luminal epithelial cells. In adult skin, p63 has a crucial role in the maintenance of epithelial stem cells. However, it is unclear whether p63 also has an equivalent role as a stem/progenitor cell factor in adult mammary epithelium. We show that p63 is essential in vivo for the survival and maintenance of parity-identified mammary epithelial cells (PI-MECs), a pregnancy-induced heterogeneous population that survives post-lactational involution and contain multipotent progenitors that give rise to alveoli and ducts in subsequent pregnancies. p63+/- glands are normal in virgin, pregnant and lactating states. Importantly, however, during the apoptotic phase of post-lactational involution p63+/- glands show a threefold increase in epithelial cell death, concomitant with increased activation of the oncostatin M/Stat3 and p53 pro-apoptotic pathways, which are responsible for this phase. Thus, p63 is a physiologic antagonist of these pathways specifically in this regressive stage. After the restructuring phase when involution is complete, mammary glands of p63+/- mice again exhibit normal epithelial architecture by conventional histology. However, using Rosa(LSL-LacZ);WAP-Cre transgenics (LSL-LacZ, lox-stop-lox β-galactosidase), a genetic in vivo labeling system for PI-MECs, we find that p63+/- glands have a 30% reduction in the number of PI-MEC progenitors and their derivatives. Importantly, PI-MECs are also cellular targets of pregnancy-promoted ErbB2 tumorigenesis. Consistent with their PI-MEC pool reduction, one-time pregnant p63+/- ErbB2 mice are partially protected from breast tumorigenesis, exhibiting extended tumor-free and overall survival, and reduced tumor multiplicity compared with their p63+/+ ErbB2 littermates. Conversely, in virgin ErbB2 mice p63 heterozygosity provides no survival advantage. In sum, our data establish that p63 is an important survival factor for pregnancy-identified PI-MEC progenitors in breast tissue in vivo.
Collapse
Affiliation(s)
- A R Yallowitz
- Department of Pathology, Stony Brook University, School of Medicine, Stony Brook, NY 11794, USA
| | - E M Alexandrova
- Department of Pathology, Stony Brook University, School of Medicine, Stony Brook, NY 11794, USA
| | - F Talos
- Department of Pathology, Stony Brook University, School of Medicine, Stony Brook, NY 11794, USA
| | - S Xu
- Department of Pathology, Stony Brook University, School of Medicine, Stony Brook, NY 11794, USA
| | - N D Marchenko
- Department of Pathology, Stony Brook University, School of Medicine, Stony Brook, NY 11794, USA
| | - U M Moll
- Department of Pathology, Stony Brook University, School of Medicine, Stony Brook, NY 11794, USA
| |
Collapse
|
47
|
Buckley NE, Nic An tSaoir CB, Blayney JK, Oram LC, Crawford NT, D’Costa ZC, Quinn JE, Kennedy RD, Harkin DP, Mullan PB. BRCA1 is a key regulator of breast differentiation through activation of Notch signalling with implications for anti-endocrine treatment of breast cancers. Nucleic Acids Res 2013; 41:8601-14. [PMID: 23863842 PMCID: PMC3794588 DOI: 10.1093/nar/gkt626] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 06/24/2013] [Accepted: 06/26/2013] [Indexed: 12/17/2022] Open
Abstract
Here, we show for the first time, that the familial breast/ovarian cancer susceptibility gene BRCA1 activates the Notch pathway in breast cells by transcriptional upregulation of Notch ligands and receptors in both normal and cancer cells. We demonstrate through chromatin immunoprecipitation assays that BRCA1 is localized to a conserved intronic enhancer region within the Notch ligand Jagged-1 (JAG1) gene, an event requiring ΔNp63. We propose that this BRCA1/ΔNp63-mediated induction of JAG1 may be important the regulation of breast stem/precursor cells, as knockdown of all three proteins resulted in increased tumoursphere growth and increased activity of stem cell markers such as Aldehyde Dehydrogenase 1 (ALDH1). Knockdown of Notch1 and JAG1 phenocopied BRCA1 knockdown resulting in the loss of Estrogen Receptor-α (ER-α) expression and other luminal markers. A Notch mimetic peptide could activate an ER-α promoter reporter in a BRCA1-dependent manner, whereas Notch inhibition using a γ-secretase inhibitor reversed this process. We demonstrate that inhibition of Notch signalling resulted in decreased sensitivity to the anti-estrogen drug Tamoxifen but increased expression of markers associated with basal-like breast cancer. Together, these findings suggest that BRCA1 transcriptional upregulation of Notch signalling is a key event in the normal differentiation process in breast tissue.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Paul B. Mullan
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, 97 Lisburn Road, Belfast BT7 9BL, UK
| |
Collapse
|
48
|
Regan JL, Sourisseau T, Soady K, Kendrick H, McCarthy A, Tang C, Brennan K, Linardopoulos S, White DE, Smalley MJ. Aurora A kinase regulates mammary epithelial cell fate by determining mitotic spindle orientation in a Notch-dependent manner. Cell Rep 2013; 4:110-23. [PMID: 23810554 DOI: 10.1016/j.celrep.2013.05.044] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 04/16/2013] [Accepted: 05/29/2013] [Indexed: 10/26/2022] Open
Abstract
Cell fate determination in the progeny of mammary epithelial stem/progenitor cells remains poorly understood. Here, we have examined the role of the mitotic kinase Aurora A (AURKA) in regulating the balance between basal and luminal mammary lineages. We find that AURKA is highly expressed in basal stem cells and, to a lesser extent, in luminal progenitors. Wild-type AURKA expression promoted luminal cell fate, but expression of an S155R mutant reduced proliferation, promoted basal fate, and inhibited serial transplantation. The mechanism involved regulation of mitotic spindle orientation by AURKA and the positioning of daughter cells after division. Remarkably, this was NOTCH dependent, as NOTCH inhibitor blocked the effect of wild-type AURKA expression on spindle orientation and instead mimicked the effect of the S155R mutant. These findings directly link AURKA, NOTCH signaling, and mitotic spindle orientation and suggest a mechanism for regulating the balance between luminal and basal lineages in the mammary gland.
Collapse
Affiliation(s)
- Joseph L Regan
- Division of Breast Cancer Research, Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Valdez JM, Zhang L, Su Q, Dakhova O, Zhang Y, Shahi P, Spencer DM, Creighton CJ, Ittmann MM, Xin L. Notch and TGFβ form a reciprocal positive regulatory loop that suppresses murine prostate basal stem/progenitor cell activity. Cell Stem Cell 2013; 11:676-88. [PMID: 23122291 DOI: 10.1016/j.stem.2012.07.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Revised: 06/04/2012] [Accepted: 07/02/2012] [Indexed: 01/17/2023]
Abstract
The role of Notch signaling in the maintenance of adult murine prostate epithelial homeostasis remains unclear. We found that Notch ligands are mainly expressed within the basal cell lineage, while active Notch signaling is detected in both the prostate basal and luminal cell lineages. Disrupting the canonical Notch effector Rbp-j impairs the differentiation of prostate basal stem cells and increases their proliferation in vitro and in vivo, but does not affect luminal cell biology. Conversely, ectopic Notch activation in adult prostates results in a decrease in basal cell number and luminal cell hyperproliferation. TGFβ dominates over Notch signaling and overrides Notch ablation-induced proliferation of prostate basal cells. However, Notch confers sensitivity and positive feedback by upregulating a plethora of TGFβ signaling components including TgfβR1. These findings reveal crucial roles of the self-enforced positive reciprocal regulatory loop between TGFβ and Notch in maintaining prostate basal stem cell dormancy.
Collapse
Affiliation(s)
- Joseph M Valdez
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Gu B, Watanabe K, Sun P, Fallahi M, Dai X. Chromatin effector Pygo2 mediates Wnt-notch crosstalk to suppress luminal/alveolar potential of mammary stem and basal cells. Cell Stem Cell 2013; 13:48-61. [PMID: 23684539 DOI: 10.1016/j.stem.2013.04.012] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 03/13/2013] [Accepted: 04/16/2013] [Indexed: 02/04/2023]
Abstract
Epigenetic mechanisms regulating lineage differentiation of mammary stem cells (MaSCs) remain poorly understood. Pygopus 2 (Pygo2) is a histone methylation reader and a context-dependent Wnt/β-catenin coactivator. Here we provide evidence for Pygo2's function in suppressing luminal/alveolar differentiation of MaSC-enriched basal cells. We show that Pygo2-deficient MaSC/basal cells exhibit partial molecular resemblance to luminal cells, such as elevated Notch signaling and reduced mammary repopulating capability upon transplantation. Inhibition of Notch signaling suppresses basal-level and Pygo2-deficiency-induced luminal/alveolar differentiation of MaSC/basal cells, whereas activation of Wnt/β-catenin signaling suppresses luminal/alveolar differentiation and Notch3 expression in a Pygo2-dependent manner. We show that Notch3 is a direct target of Pygo2 and that Pygo2 is required for β-catenin binding and maintenance of a poised/repressed chromatin state at the Notch3 locus in MaSC/basal cells. Together, our data support a model where Pygo2-mediated chromatin regulation connects Wnt signaling and Notch signaling to restrict the luminal/alveolar differentiation competence of MaSC/basal cells.
Collapse
Affiliation(s)
- Bingnan Gu
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | | | | | | | | |
Collapse
|