51
|
Chen JQ, Mori H, Cardiff RD, Trott JF, Hovey RC, Hubbard NE, Engelberg JA, Tepper CG, Willis BJ, Khan IH, Ravindran RK, Chan SR, Schreiber RD, Borowsky AD. Abnormal Mammary Development in 129:STAT1-Null Mice is Stroma-Dependent. PLoS One 2015; 10:e0129895. [PMID: 26075897 PMCID: PMC4468083 DOI: 10.1371/journal.pone.0129895] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 05/14/2015] [Indexed: 11/18/2022] Open
Abstract
Female 129:Stat1-null mice (129S6/SvEvTac-Stat1tm1Rds homozygous) uniquely develop estrogen-receptor (ER)-positive mammary tumors. Herein we report that the mammary glands (MG) of these mice have altered growth and development with abnormal terminal end buds alongside defective branching morphogenesis and ductal elongation. We also find that the 129:Stat1-null mammary fat pad (MFP) fails to sustain the growth of 129S6/SvEv wild-type and Stat1-null epithelium. These abnormalities are partially reversed by elevated serum progesterone and prolactin whereas transplantation of wild-type bone marrow into 129:Stat1-null mice does not reverse the MG developmental defects. Medium conditioned by 129:Stat1-null epithelium-cleared MFP does not stimulate epithelial proliferation, whereas it is stimulated by medium conditioned by epithelium-cleared MFP from either wild-type or 129:Stat1-null females having elevated progesterone and prolactin. Microarrays and multiplexed cytokine assays reveal that the MG of 129:Stat1-null mice has lower levels of growth factors that have been implicated in normal MG growth and development. Transplanted 129:Stat1-null tumors and their isolated cells also grow slower in 129:Stat1-null MG compared to wild-type recipient MG. These studies demonstrate that growth of normal and neoplastic 129:Stat1-null epithelium is dependent on the hormonal milieu and on factors from the mammary stroma such as cytokines. While the individual or combined effects of these factors remains to be resolved, our data supports the role of STAT1 in maintaining a tumor-suppressive MG microenvironment.
Collapse
Affiliation(s)
- Jane Q. Chen
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Hidetoshi Mori
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Robert D. Cardiff
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Josephine F. Trott
- Department of Animal Science, University of California, Davis, California, United States of America
| | - Russell C. Hovey
- Department of Animal Science, University of California, Davis, California, United States of America
| | - Neil E. Hubbard
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Jesse A. Engelberg
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Clifford G. Tepper
- Division of Basic Sciences, Cancer Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Sacramento, California, United States of America
| | - Brandon J. Willis
- Mouse Biology Program, University of California, Davis, California, United States of America
| | - Imran H. Khan
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Resmi K. Ravindran
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Szeman R. Chan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Robert D. Schreiber
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Alexander D. Borowsky
- Center for Comparative Medicine, University of California, Davis, California, United States of America
- Department of Pathology and Laboratory Medicine, University of California, Davis, School of Medicine, Sacramento, California, United States of America
- * E-mail:
| |
Collapse
|
52
|
Zaragozá R, García-Trevijano ER, Lluch A, Ribas G, Viña JR. Involvement of Different networks in mammary gland involution after the pregnancy/lactation cycle: Implications in breast cancer. IUBMB Life 2015; 67:227-38. [PMID: 25904072 DOI: 10.1002/iub.1365] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 02/03/2015] [Indexed: 11/06/2022]
Abstract
Early pregnancy is associated with a reduction in a woman's lifetime risk for breast cancer. However, different studies have demonstrated an increase in breast cancer risk in the years immediately following pregnancy. Early and long-term risk is even higher if the mother age is above 35 years at the time of first parity. The proinflammatory microenvironment within the mammary gland after pregnancy renders an "ideal niche" for oncogenic events. Signaling pathways involved in programmed cell death and tissue remodeling during involution are also activated in breast cancer. Herein, the major signaling pathways involved in mammary gland involution, signal transducer and activator of transcription (STAT3), nuclear factor-kappa B (NF-κB), transforming growth factor beta (TGFβ), and retinoid acid receptors (RARs)/retinoid X receptors (RXRs), are reviewed as part of the complex network of signaling pathways that crosstalk in a contextual-dependent manner. These factors, also involved in breast cancer development, are important regulatory nodes for signaling amplification after weaning. Indeed, during involution, p65/p300 target genes such as MMP9, Capn1, and Capn2 are upregulated. Elevated expression and activities of these proteases in breast cancer have been extensively documented. The role of these proteases during mammary gland involution is further discussed. MMPs, calpains, and cathepsins exert their effect by modification of the extracellular matrix and intracellular proteins. Calpains, activated in the mammary gland during involution, cleave several proteins located in cell membrane, lysosomes, mitochondria, and nuclei favoring cell death. Besides, during this period, Capn1 is most probably involved in the modulation of preadipocyte differentiation through chromatin remodeling. Calpains can be implicated in cell anchoring loss, providing a proper microenvironment for tumor growth. A better understanding of the role of any of these proteases in tumorigenesis may yield novel therapeutic targets or prognostic markers for breast cancer.
Collapse
Affiliation(s)
- Rosa Zaragozá
- Instituto INCLIVA, Facultad de Medicina/Hospital Clínico, Universidad de Valencia, Valencia, Spain
| | - Elena R García-Trevijano
- Instituto INCLIVA, Facultad de Medicina/Hospital Clínico, Universidad de Valencia, Valencia, Spain.,Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Ana Lluch
- Instituto INCLIVA, Facultad de Medicina/Hospital Clínico, Universidad de Valencia, Valencia, Spain.,Servicio Oncología Médica, Hospital Clínico Universitario Valencia, Valencia, Spain
| | - Gloria Ribas
- Instituto INCLIVA, Facultad de Medicina/Hospital Clínico, Universidad de Valencia, Valencia, Spain.,Servicio Oncología Médica, Hospital Clínico Universitario Valencia, Valencia, Spain
| | - Juan R Viña
- Instituto INCLIVA, Facultad de Medicina/Hospital Clínico, Universidad de Valencia, Valencia, Spain.,Servicio Oncología Médica, Hospital Clínico Universitario Valencia, Valencia, Spain
| |
Collapse
|
53
|
Khaled WT, Choon Lee S, Stingl J, Chen X, Raza Ali H, Rueda OM, Hadi F, Wang J, Yu Y, Chin SF, Stratton M, Futreal A, Jenkins NA, Aparicio S, Copeland NG, Watson CJ, Caldas C, Liu P. BCL11A is a triple-negative breast cancer gene with critical functions in stem and progenitor cells. Nat Commun 2015; 6:5987. [PMID: 25574598 PMCID: PMC4338552 DOI: 10.1038/ncomms6987] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 11/28/2014] [Indexed: 01/03/2023] Open
Abstract
Triple-negative breast cancer (TNBC) has poor prognostic outcome compared with other types of breast cancer. The molecular and cellular mechanisms underlying TNBC pathology are not fully understood. Here, we report that the transcription factor BCL11A is overexpressed in TNBC including basal-like breast cancer (BLBC) and that its genomic locus is amplified in up to 38% of BLBC tumours. Exogenous BCL11A overexpression promotes tumour formation, whereas its knockdown in TNBC cell lines suppresses their tumourigenic potential in xenograft models. In the DMBA-induced tumour model, Bcl11a deletion substantially decreases tumour formation, even in p53-null cells and inactivation of Bcl11a in established tumours causes their regression. At the cellular level, Bcl11a deletion causes a reduction in the number of mammary epithelial stem and progenitor cells. Thus, BCL11A has an important role in TNBC and normal mammary epithelial cells. This study highlights the importance of further investigation of BCL11A in TNBC-targeted therapies.
Collapse
Affiliation(s)
- Walid T. Khaled
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1HH, UK
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, UK
- These authors contributed equally to this work
| | - Song Choon Lee
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1HH, UK
- These authors contributed equally to this work
| | - John Stingl
- Cancer Research UK Cambridge Institute, and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Xiongfeng Chen
- SAIC-Frederic, National Cancer Institute-Frederick, Frederick, Maryland 21701, USA
| | - H. Raza Ali
- Cancer Research UK Cambridge Institute, and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
- Cambridge Experimental Cancer Medicine Centre, Cambridge CB2 0RE, UK
| | - Oscar M. Rueda
- Cancer Research UK Cambridge Institute, and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Fazal Hadi
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, UK
| | - Juexuan Wang
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1HH, UK
| | - Yong Yu
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1HH, UK
| | - Suet-Feung Chin
- Cancer Research UK Cambridge Institute, and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Mike Stratton
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1HH, UK
| | - Andy Futreal
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1HH, UK
| | - Nancy A. Jenkins
- The Methodist Hospital Research Institute, 6670 Bertner Street, Houston, Texas 77030, USA
| | - Sam Aparicio
- Molecular Oncology Department, BC Cancer Agency Research Centre, 675 West 10th Avenue, Vancouver, British Columbia V5Z 1L3, Canada
| | - Neal G. Copeland
- The Methodist Hospital Research Institute, 6670 Bertner Street, Houston, Texas 77030, USA
| | | | - Carlos Caldas
- Cancer Research UK Cambridge Institute, and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
- Cambridge Experimental Cancer Medicine Centre, Cambridge CB2 0RE, UK
- Addenbrooke’s Hospital, Cambridge University Hospital NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge CB2 2QQ, UK
| | - Pentao Liu
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1HH, UK
| |
Collapse
|
54
|
von Moltke J, Locksley RM. I-L-C-2 it: type 2 immunity and group 2 innate lymphoid cells in homeostasis. Curr Opin Immunol 2014; 31:58-65. [PMID: 25458996 DOI: 10.1016/j.coi.2014.09.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/24/2014] [Accepted: 09/28/2014] [Indexed: 12/17/2022]
Abstract
Innate type 2 immune cells are activated in response to helminths, allergens, and certain types of proteases and particulates. Recently, innate type 2 immune pathways have also been implicated in protective host responses to homeostatic perturbations, such as metabolic dysfunction, atherosclerosis, and tissue injury. In this context, innate type 2 cytokines stimulate local tissues, recruit eosinophils, and alternatively activate macrophages to restore homeostasis. As the major source of innate interleukin (IL)-5 and IL-13, group 2 innate lymphoid cells are positioned to initiate and maintain homeostatic type 2 responses. The absence of exogenous stimuli in these processes implicates endogenous pathways in the activation of type 2 immunity and suggests an alternative evolutionary trajectory for type 2 immunity, apart from its role in response to helminths and allergens.
Collapse
Affiliation(s)
- Jakob von Moltke
- Departments of Microbiology and Immunology and Medicine, Howard Hughes Medical Institute, University of California, San Francisco, CA 94143, USA
| | - Richard M Locksley
- Departments of Microbiology and Immunology and Medicine, Howard Hughes Medical Institute, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
55
|
Pensa S, Lloyd-Lewis B, Sargeant TJ, Resemann HK, Kahn CR, Watson CJ. Signal transducer and activator of transcription 3 and the phosphatidylinositol 3-kinase regulatory subunits p55α and p50α regulate autophagy in vivo. FEBS J 2014; 281:4557-67. [PMID: 25205393 DOI: 10.1111/febs.13035] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 08/11/2014] [Accepted: 09/02/2014] [Indexed: 02/04/2023]
Abstract
Mammary gland involution involves a process that includes one of the most dramatic examples of cell death in an adult mammalian organism. We have previously shown that signal transducer and activator of transcription 3 (Stat3) regulates a lysosomal pathway of cell death in the first 48 h of involution and induces lysosome leakiness in mammary epithelial cells. Interestingly, Stat3 is associated also with the striking induction of autophagy that occurs concomitantly with cell death, presumably as a transient survival mechanism. The phosphatidylinositol 3-kinase regulatory subunits p55α and p50α are dramatically and specifically upregulated at the transcriptional level by Stat3 at the onset of involution. We show here that ablation of either Stat3 or p55α/p50α in vivo affects autophagy during involution. We used two different cell culture models (normal mammary epithelial cells and mouse embryonic fibroblasts) to further investigate the role of p55α/p50α in autophagy regulation. Our results demonstrate a direct role for p55α/p50α as inhibitors of autophagy mediated by p85α. Thus, Stat3 and its downstream targets p55α/p50α are key regulators of the balance between autophagy and cell death in vivo.
Collapse
Affiliation(s)
- Sara Pensa
- Department of Pathology, University of Cambridge, Cambridge, UK
| | | | | | | | | | | |
Collapse
|
56
|
Hormonal regulation of the cytokine microenvironment in the mammary gland. J Reprod Immunol 2014; 106:58-66. [PMID: 25138705 DOI: 10.1016/j.jri.2014.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/04/2014] [Accepted: 07/05/2014] [Indexed: 11/22/2022]
Abstract
The mammary gland is a unique organ that undergoes hormone-driven developmental changes over the course of the ovarian cycle during adult life. Macrophages play a role in regulating cellular turnover in the mammary gland and may affect cancer susceptibility. However, the immune microenvironment that regulates macrophage function has not been described. Hormonal regulation of the cytokine microenvironment across the ovarian cycle was explored using microbead multiplex assay for 15 cytokines in mammary glands from C57Bl/6 mice at different stages of the oestrous cycle, and in ovariectomised mice administered oestradiol and progesterone. The cytokines that were found to fluctuate over the course of the oestrous cycle were colony-stimulating factor (CSF)1, CSF2, interferon gamma (IFNG) and tumour necrosis factor alpha (TNFA), all of which were significantly elevated at oestrus compared with other phases. The concentration of serum progesterone during the oestrus phase negatively correlated with the abundance of cytokines CSF3, IL12p40, IFNG and leukaemia inhibitory factor (LIF). In ovariectomised mice, exogenous oestradiol administration increased mammary gland CSF1, CSF2, IFNG and LIF, compared with ovariectomised control mice. Progesterone administration together with oestradiol resulted in reduced CSF1, CSF3 and IFNG compared with oestradiol administration alone. This study suggests that the cytokine microenvironment in the mammary gland at the oestrus phase of the ovarian cycle is relatively pro-inflammatory compared with other stages of the cycle, and that the oestradiol-induced cytokine microenvironment is significantly attenuated by progesterone. A continuously fluctuating cytokine microenvironment in the mammary gland presumably regulates the phenotypes of resident leukocytes and may affect mammary gland cancer susceptibility.
Collapse
|
57
|
Sun X, Ingman WV. Cytokine networks that mediate epithelial cell-macrophage crosstalk in the mammary gland: implications for development and cancer. J Mammary Gland Biol Neoplasia 2014; 19:191-201. [PMID: 24924120 DOI: 10.1007/s10911-014-9319-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 05/19/2014] [Indexed: 01/28/2023] Open
Abstract
Dynamic interactions between the hormone responsive mammary gland epithelium and surrounding stromal macrophage populations are critical for normal development and function of the mammary gland. Macrophages are versatile cells capable of diverse roles in mammary gland development and maintenance of homeostasis, and their function is highly dependent on signals within the local cytokine microenvironment. The mammary epithelium secretes a number of cytokines, including colony stimulating factor 1 (CSF1), transforming growth factor beta 1 (TGFB1), and chemokine ligand 2 (CCL2) that affect the abundance, phenotype and function of macrophages. However, aberrations in these interactions have been found to increase the risk of tumour formation, and utilisation of stromal macrophage support by tumours can increase the invasive and metastatic potential of the cancer. Studies utilising genetically modified mouse models have shed light on the significance of epithelial cell-macrophage crosstalk, and the cytokines that mediate this communication, in mammary gland development and tumourigenesis. This article reviews the current status of our understanding of the roles of epithelial cell-derived cytokines in mammary gland development and cancer, with a focus on the crosstalk between epithelial cells and the local macrophage population.
Collapse
Affiliation(s)
- Xuan Sun
- School of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, Australia
| | | |
Collapse
|
58
|
Need EF, Atashgaran V, Ingman WV, Dasari P. Hormonal regulation of the immune microenvironment in the mammary gland. J Mammary Gland Biol Neoplasia 2014; 19:229-39. [PMID: 24993978 DOI: 10.1007/s10911-014-9324-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 06/16/2014] [Indexed: 12/29/2022] Open
Abstract
It is well established that the development and homeostasis of the mammary gland are highly dependent upon the actions of ovarian hormones progesterone and estrogen, as well as the availability of prolactin for the pregnant and lactating gland. More recently it has become apparent that immune system cells and cytokines play essential roles in both mammary gland development as well as breast cancer. Here, we review hormonal effects on mammary gland biology during puberty, menstrual cycling, pregnancy, lactation and involution, and dissect how hormonal control of the immune system may contribute to mammary development at each stage via cytokine secretion and recruitment of macrophages, eosinophils, mast cells and lymphocytes. Collectively, these alterations may create an immunotolerant or inflammatory immune environment at specific developmental stages or phases of the menstrual cycle. Of particular interest for further research is investigation of the combinatorial actions of progesterone and estrogen during the luteal phase of the menstrual cycle and key developmental points where the immune system may play an active role both in mammary development as well as in the creation of an immunotolerant environment, thereby affecting breast cancer risk.
Collapse
Affiliation(s)
- Eleanor F Need
- Discipline of Surgery, School of Medicine, The Queen Elizabeth Hospital, University of Adelaide, DX465702, 28 Woodville Road, Woodville, SA, 5011, Australia
| | | | | | | |
Collapse
|
59
|
Hughes K, Watson CJ. The spectrum of STAT functions in mammary gland development. JAKSTAT 2014; 1:151-8. [PMID: 24058764 PMCID: PMC3670238 DOI: 10.4161/jkst.19691] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 02/08/2012] [Accepted: 02/13/2012] [Indexed: 12/21/2022] Open
Abstract
The signal transducer and activator of transcription (STAT) family of transcription factors have a spectrum of functions in mammary gland development. In some cases these roles parallel those of STATs in other organ systems, while in other instances the function of individual STATs in the mammary gland is specific to this tissue. In the immune system, STAT6 is associated with differentiation of T helper cells, while in the mammary gland, it has a fundamental role in the commitment of luminal epithelial cells to the alveolar lineage. STAT5A is required for the production of luminal progenitor cells from mammary stem cells and is essential for the differentiation of milk producing alveolar cells during pregnancy. By contrast, the initiation of regression following weaning heralds a dramatic and specific activation of STAT3, reflecting its pivotal role in the regulation of cell death and tissue remodeling during mammary involution. Although it has been demonstrated that STAT1 is regulated during a mammary developmental cycle, it is not yet determined whether it has a specific, non-redundant function. Thus, the mammary gland constitutes an unusual example of an adult organ in which different STATs are sequentially activated to orchestrate the processes of functional differentiation, cell death and tissue remodeling.
Collapse
|
60
|
The PI3K regulatory subunits p55α and p50α regulate cell death in vivo. Cell Death Differ 2014; 21:1442-50. [PMID: 24902901 DOI: 10.1038/cdd.2014.59] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 02/10/2014] [Accepted: 02/21/2014] [Indexed: 02/07/2023] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K) regulatory subunits p55α and p50α are coordinately transcriptionally upregulated by signal transducer and activator of transcription 3 (Stat3) at the onset of mammary gland involution, a process that requires Stat3. Deletion of both p55α and p50α subunits in vivo abrogated mammary epithelial cell death during involution. This was associated also with reduced cytosolic levels and activity of the cysteine protease cathepsin L, which is implicated in lysosomal-mediated programmed cell death (LM-PCD) and is upregulated in involution. Furthermore, involution is delayed in cathepsin L-deficient mice suggesting that the p55α/p50α subunits mediate cell death in part by elevating the level of cathepsin L resulting in increased cytosolic activity. Surprisingly, we found that p55α/p50α localize to the nucleus where they bind to chromatin and regulate transcription of a subset of inflammatory/acute phase genes that are also Stat3 targets. Our findings reveal a novel role for these PI3K regulatory subunits as regulators of LM-PCD in vivo.
Collapse
|
61
|
Haricharan S, Li Y. STAT signaling in mammary gland differentiation, cell survival and tumorigenesis. Mol Cell Endocrinol 2014; 382:560-569. [PMID: 23541951 PMCID: PMC3748257 DOI: 10.1016/j.mce.2013.03.014] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 03/18/2013] [Indexed: 01/10/2023]
Abstract
The mammary gland is a unique organ that undergoes extensive and profound changes during puberty, menstruation, pregnancy, lactation and involution. The changes that take place during puberty involve large-scale proliferation and invasion of the fat-pad. During pregnancy and lactation, the mammary cells are exposed to signaling pathways that inhibit apoptosis, induce proliferation and invoke terminal differentiation. Finally, during involution the mammary gland is exposed to milk stasis, programmed cell death and stromal reorganization to clear the differentiated milk-producing cells. Not surprisingly, the signaling pathways responsible for bringing about these changes in breast cells are often subverted during the process of tumorigenesis. The STAT family of proteins is involved in every stage of mammary gland development, and is also frequently implicated in breast tumorigenesis. While the roles of STAT3 and STAT5 during mammary gland development and tumorigenesis are well studied, others members, e.g. STAT1 and STAT6, have only recently been observed to play a role in mammary gland biology. Continued investigation into the STAT protein network in the mammary gland will likely yield new biomarkers and risk factors for breast cancer, and may also lead to novel prophylactic or therapeutic strategies against breast cancer.
Collapse
Affiliation(s)
- S Haricharan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Y Li
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
62
|
Campbell JJ, Botos LA, Sargeant TJ, Davidenko N, Cameron RE, Watson CJ. A 3-D in vitro co-culture model of mammary gland involution. Integr Biol (Camb) 2014; 6:618-26. [DOI: 10.1039/c3ib40257f] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
An in vitro model of mammary gland supporting 3D cell–cell and cell–matrix interactions demonstrates complete in vivo-like neo-tissue formation and remodelling processes (involution) under hormonal control.
Collapse
Affiliation(s)
| | | | | | | | - Ruth E. Cameron
- Department of Materials Science and Metallurgy
- Cambridge CB3 0FS, UK
| | | |
Collapse
|
63
|
Haricharan S, Dong J, Hein S, Reddy JP, Du Z, Toneff M, Holloway K, Hilsenbeck SG, Huang S, Atkinson R, Woodward W, Jindal S, Borges VF, Gutierrez C, Zhang H, Schedin PJ, Osborne CK, Tweardy DJ, Li Y. Mechanism and preclinical prevention of increased breast cancer risk caused by pregnancy. eLife 2013; 2:e00996. [PMID: 24381245 PMCID: PMC3874103 DOI: 10.7554/elife.00996] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
While a first pregnancy before age 22 lowers breast cancer risk, a pregnancy after age 35 significantly increases life-long breast cancer risk. Pregnancy causes several changes to the normal breast that raise barriers to transformation, but how pregnancy can also increase cancer risk remains unclear. We show in mice that pregnancy has different effects on the few early lesions that have already developed in the otherwise normal breast—it causes apoptosis evasion and accelerated progression to cancer. The apoptosis evasion is due to the normally tightly controlled STAT5 signaling going astray—these precancerous cells activate STAT5 in response to pregnancy/lactation hormones and maintain STAT5 activation even during involution, thus preventing the apoptosis normally initiated by oncoprotein and involution. Short-term anti-STAT5 treatment of lactation-completed mice bearing early lesions eliminates the increased risk after a pregnancy. This chemoprevention strategy has important implications for preventing increased human breast cancer risk caused by pregnancy. DOI:http://dx.doi.org/10.7554/eLife.00996.001 Pregnancy changes the probability that a woman will later develop breast cancer. If a woman’s first pregnancy occurs before her 22nd birthday, the chances of developing breast cancer are reduced. However, if the first pregnancy occurs after her 35th birthday, there is an increased risk of breast cancer. It is not clear why this age-related difference exists, but as more women wait until their 30s to start a family, there is greater urgency to understand this difference. Breasts undergo extensive changes during pregnancy. This remodeling makes their cells less likely to multiply, and also less likely to develop tumors, which could explain the protective effect of pregnancy for younger women. But why would older women not reap the same benefits? One hypothesis is that older first-time mothers are more likely than younger first-time mothers to already have breast tissue with cells carrying cancer-causing mutations, or to have clusters of abnormal precancerous cells. Now, Haricharan et al. have tested this hypothesis by inserting two cancer-causing genes into female mice. Half of the mice were then made pregnant and allowed to nurse their young, whilst the other half were never mated. Although, both groups of mice later developed tumors, the mice that had been pregnant developed more tumors and did so faster. The increased cancer levels in the mice that had been pregnant were not due to them having more precancerous cells at the early stages of pregnancy than the unmated mice of the same age. Further, the precancerous cells in the impregnated mice did not proliferate faster than those in the mice that were never pregnant. Instead, pregnancy weakened the protective process that culls pre-existing precancerous cells. These cells evaded destruction by activating a signaling pathway called the STAT5 pathway in response to pregnancy hormones. Haricharan et al. also examined tissue samples from women with a very early form of breast cancer and found elevated levels of STAT5 in tumors from women who had been pregnant compared to those who had not been pregnant. The good news is that precancerous cells do not always become cancerous. However, for those women with a high risk of developing breast cancer, Haricharan et al. suggest that temporarily reducing STAT5 activity after pregnancy with medication might reduce this risk. Treating mice with anti-STAT5 drugs for a few weeks after they finished nursing their young lessened the elevated cancer risk, and so the next challenge is to see if this approach will also be effective in human clinical trials. DOI:http://dx.doi.org/10.7554/eLife.00996.002
Collapse
Affiliation(s)
- Svasti Haricharan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Therapeutic modulators of STAT signalling for human diseases. Nat Rev Drug Discov 2013; 12:611-29. [PMID: 23903221 DOI: 10.1038/nrd4088] [Citation(s) in RCA: 344] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The signal transducer and activator of transcription (STAT) proteins have important roles in biological processes. The abnormal activation of STAT signalling pathways is also implicated in many human diseases, including cancer, autoimmune diseases, rheumatoid arthritis, asthma and diabetes. Over a decade has passed since the first inhibitor of a STAT protein was reported and efforts to discover modulators of STAT signalling as therapeutics continue. This Review discusses the outcomes of the ongoing drug discovery research endeavours against STAT proteins, provides perspectives on new directions for accelerating the discovery of drug candidates, and highlights the noteworthy candidate therapeutics that have progressed to clinical trials.
Collapse
|
65
|
Csanaky K, Doppler W, Tamas A, Kovacs K, Toth G, Reglodi D. Influence of terminal differentiation and PACAP on the cytokine, chemokine, and growth factor secretion of mammary epithelial cells. J Mol Neurosci 2013; 52:28-36. [PMID: 24323361 DOI: 10.1007/s12031-013-0193-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 11/21/2013] [Indexed: 11/27/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP), a neuropeptide with trophic and cytoprotective effects, has been shown to affect cell survival, proliferation, and also differentiation of various cell types. The high PACAP level in the milk and its changes during lactation suggest a possible effect of PACAP on the differentiation of mammary epithelial cells. Mammary cell differentiation is regulated by hormones, growth factors, cytokines/chemokines, and angiogenic proteins. In this study, differentiation was hormonally induced by lactogenic hormones in confluent cultures of HC11 mouse mammary epithelial cells. We investigated the effect of PACAP on mammary cell differentiation as well as release of cytokines, chemokines, and growth factors. Differentiation was assessed by expression analysis of the milk protein β-casein. Differentiation significantly decreased the secretion of interferon gammainduced protein (IP)-10, "regulated upon activation normal T cell expressed and presumably secreted" (RANTES), insulin-like growth factor-binding protein (IGFBP)-3 and the epidermal growth factor receptor (EGFR) ligands, such as epidermal growth factor (EGF) and amphiregulin (AREG). The changes in the levels of IP-10 and RANTES may be relevant for the alterations in homing of T cells and B cells at different stages of mammary gland development, while the changes of the EGFR ligands may facilitate the switch from proliferative to lactating stage. PACAP did not modulate the expression of β-casein or the activity of hormone-induced pathways as determined by the analysis of phosphorylation of Akt, STAT5, and p38 MAPK. However, PACAP decreased the release of EGF and AREG from non-differentiated cells. This may influence the extracellular signal-related transactivation of EGFR in the non-differentiated mammary epithelium and is considered to have an impact on the modulation of oncogenic EGFR signaling in breast cancer.
Collapse
Affiliation(s)
- Katalin Csanaky
- Department of Anatomy, PTE-MTA "Lendulet" PACAP Research Team, University of Pecs, Szigeti ut 12, Pecs, 7624, Hungary
| | | | | | | | | | | |
Collapse
|
66
|
Haricharan S, Hein SM, Dong J, Toneff MJ, Aina OH, Rao PH, Cardiff RD, Li Y. Contribution of an alveolar cell of origin to the high-grade malignant phenotype of pregnancy-associated breast cancer. Oncogene 2013; 33:5729-39. [PMID: 24317513 PMCID: PMC4050040 DOI: 10.1038/onc.2013.521] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 10/17/2013] [Accepted: 10/23/2013] [Indexed: 12/15/2022]
Abstract
Pregnancy-associated breast cancers (PABCs) are tumors diagnosed during pregnancy or up to 5 years following parturition, and are usually high-grade, connective tissue-rich, and estrogen receptor (ER)/progesterone receptor-negative. Little is known about the cellular origin of PABCs or the mechanisms by which PABCs are initiated. Using the RCAS retrovirus to deliver the ErbB2 oncogene into the mammary epithelium of our previously reported MMTV-tva transgenic mice, we detected high-grade, poorly differentiated, stroma-rich and ER-negative tumors during pregnancy and lactation. These high-grade and stroma-rich tumors were less frequent in involuted mice or in age-matched nulliparous mice. More importantly, by generating a WAP-tva transgenic line for expression of ErbB2 selectively in WAP(+) mammary alveolar cells, we found that tumors had similar morphological phenotypes (high grade, poorly differentiated, stroma-rich and ER-negative), irrespective of the time since pregnancy and even in the absence of pregnancy. These data suggest that PABCs arise preferentially from an alveolar cell population that expands during pregnancy and lactation. This somatic mouse model may also be useful for preclinical testing of new prophylactic and therapeutic strategies against PABC.
Collapse
Affiliation(s)
- S Haricharan
- Lester & Sue Smith Breast Center and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - S M Hein
- Lester & Sue Smith Breast Center and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - J Dong
- Lester & Sue Smith Breast Center and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - M J Toneff
- Lester & Sue Smith Breast Center and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - O H Aina
- Center for Comparative Medicine, University of California at Davis, Davis, CA, USA
| | - P H Rao
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, USA
| | - R D Cardiff
- Center for Comparative Medicine, University of California at Davis, Davis, CA, USA
| | - Y Li
- Lester & Sue Smith Breast Center and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
67
|
Hodson LJ, Chua AC, Evdokiou A, Robertson SA, Ingman WV. Macrophage Phenotype in the Mammary Gland Fluctuates over the Course of the Estrous Cycle and Is Regulated by Ovarian Steroid Hormones1. Biol Reprod 2013; 89:65. [DOI: 10.1095/biolreprod.113.109561] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
68
|
Oliver CH, Nichols J, Watson CJ. The KRAB domain zinc finger protein, Zfp157, is expressed in multiple tissues during mouse embryogenesis and in specific cells in adult mammary gland and skin. Genesis 2013; 51:179-86. [PMID: 23315963 DOI: 10.1002/dvg.22367] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 11/23/2012] [Accepted: 01/03/2013] [Indexed: 11/07/2022]
Abstract
The functions of members of the large family of transcriptional repressors, the KRAB domain zinc finger proteins, are not well described. We have identified a new member of this family, Zfp157, as a downstream target of the transcription factor Stat6 in mammary gland. Using a gene-trap approach, we have generated mice harboring a Zfp157-LacZ reporter gene. We have characterized the expression of this reporter during mouse embryogenesis and show that it is expressed in the epiblast and subsequently in a number of embryonic tissues including brain, ovary, intestine, kidney, lung, mammary gland, and hair follicle. In the adult, Zfp157 continues to be expressed in a wide range of tissues while specific patterns of reporter gene expression are apparent in the mammary gland, primarily in the basal epithelial cells of ducts and in the sebaceous glands of hair follicles. These data lay the foundation for further work on the function of Zfp157.
Collapse
Affiliation(s)
- Carrie H Oliver
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, United Kingdom
| | | | | |
Collapse
|
69
|
Abstract
Mammary glands are crucial to the reproductive strategy of mammals, and the milk of domesticated ruminants serves as an important source of nutrients for the human population. The majority of mammary gland development occurs postnatally, and the mammary gland undergoes cyclical periods of growth, differentiation, lactation, and regression that are coordinated to provide nutrients for offspring or are driven by strategies to manage reproduction and milk production of domesticated species. Growth and maintenance of the mammary epithelium depends on the function of mammary stem cells and progenitor cells. In this review, we provide an overview of postnatal mammary gland development, cyclical phases of mammary gland regression (regression during lactation and between successive lactations), and mammary stem cells and progenitor cells. Where possible, these processes are related to animal production and compared across species, particularly bovine, porcine, murine, and human.
Collapse
Affiliation(s)
- Anthony V Capuco
- Bovine Functional Genomics Laboratory, US Department of Agriculture, Agricultural Research Service, Beltsville, Maryland 20705;
| | | |
Collapse
|
70
|
Hallett MA, Venmar KT, Fingleton B. Cytokine stimulation of epithelial cancer cells: the similar and divergent functions of IL-4 and IL-13. Cancer Res 2012; 72:6338-43. [PMID: 23222300 DOI: 10.1158/0008-5472.can-12-3544] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The Th2 cytokines interleukin (IL)-4 and -13 are acknowledged regulators of lymphocyte proliferation and activation. They have also been well studied in the regulation of various myeloid-derived populations in tumor biology. It has become clear, however, that both cytokines can have direct effects on epithelial tumor cells expressing appropriate receptors. Changes in tumor proliferation, survival, and metastatic capability have all been ascribed to IL-4 and/or IL-13 action. Here, we evaluate the evidence to support direct tumor-promoting roles of these cytokines. We also identify the questions that should be addressed before proceeding with therapeutic approaches based on neutralization of IL-4 or IL-13 pathways.
Collapse
Affiliation(s)
- Miranda A Hallett
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | | |
Collapse
|
71
|
Yu Z, Pestell TG, Lisanti MP, Pestell RG. Cancer stem cells. Int J Biochem Cell Biol 2012; 44:2144-51. [PMID: 22981632 PMCID: PMC3496019 DOI: 10.1016/j.biocel.2012.08.022] [Citation(s) in RCA: 495] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 08/06/2012] [Accepted: 08/27/2012] [Indexed: 12/13/2022]
Abstract
Cancer stem cells (CSCs) are a small subpopulation of cells within tumors with capabilities of self-renewal, differentiation, and tumorigenicity when transplanted into an animal host. A number of cell surface markers such as CD44, CD24, and CD133 are often used to identify and enrich CSCs. A regulatory network consisting of microRNAs and Wnt/β-catenin, Notch, and Hedgehog signaling pathways controls CSC properties. The clinical relevance of CSCs has been strengthened by emerging evidence, demonstrating that CSCs are resistant to conventional chemotherapy and radiation treatment and that CSCs are very likely to be the origin of cancer metastasis. CSCs are believed to be an important target for novel anti-cancer drug discovery. Herein we summarize the current understanding of CSCs, with a focus on the role of miRNA and epithelial-mesenchymal transition (EMT), and discuss the clinical application of targeting CSCs for cancer treatment.
Collapse
Affiliation(s)
- Zuoren Yu
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Zuoren Yu, Ph.D. Research Center for Translational Medicine East Hospital, Tongji University School of Medicine Shanghai 200120 China
| | - Timothy G. Pestell
- Department of Stem Cell Biology and Regenerative Medicine, Kimmel Cancer Center, Thomas Jefferson University, 233 South 10 St. Philadelphia PA 19107
| | - Michael P. Lisanti
- Department of Stem Cell Biology and Regenerative Medicine, Kimmel Cancer Center, Thomas Jefferson University, 233 South 10 St. Philadelphia PA 19107
| | - Richard G. Pestell
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, 233 South 10 St. Philadelphia PA 19107
- Correspondence to: Richard G. Pestell, M.D., Ph.D. Department of Cancer Biology Thomas Jefferson University Kimmel Cancer Center 233 S 10th Street, Suite 1050 Philadelphia, PA 19107 Tel: 215-503-5692, Fax: 215-503-9334
| |
Collapse
|
72
|
Brenaut P, Bangera R, Bevilacqua C, Rebours E, Cebo C, Martin P. Validation of RNA isolated from milk fat globules to profile mammary epithelial cell expression during lactation and transcriptional response to a bacterial infection. J Dairy Sci 2012; 95:6130-44. [PMID: 22921620 DOI: 10.3168/jds.2012-5604] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 05/22/2012] [Indexed: 12/12/2022]
Abstract
Mastitis, an inflammation of the mammary gland, is the most costly infectious disease of dairy ruminants worldwide. Although it receives considerable attention, the early steps of the host response remain poorly defined. Here, we report a noninvasive method using milk fat globules (MFG) as a source of mammary RNA to follow the dynamics of the global transcriptional response of mammary epithelial cells (MEC) during the course of a bacterial infection. We first assessed that RNA isolated from MFG were representative of MEC RNA; we then evaluated whether MFG RNA could be used to monitor the MEC response to infection. Sufficiently high yields of good-quality RNA (RNA integrity numbers ranging between 6.7 and 8.7) were obtained from goat MFG for subsequent analyses. Contamination of MFG by macrophages and neutrophils, which can be trapped during creaming, was assessed and when using quantitative real-time PCR for cell-type specific markers, was shown to be weak enough (<8%) to affect MFG gene expression profiling. Using microarrays, we showed that RNA extracted from MFG and from mammary alveolar parenchyma shared approximately 90% of the highlighted probes corresponding in particular to genes encoding milk proteins (CSN, BLG, LALBA) and enzymes involved in milk fat synthesis and secretion (FASN, XDH, ADRP, SCD, and DGAT1). In addition, a gene involved in the acute-phase reaction, coding for the serum amyloid A3 (SAA3) protein, was found within the first 50 most highly expressed genes in a noninfectious context in both mammary alveolar parenchyma and MFG, strongly suggesting that SAA3 is expressed in MEC. We took advantage of this noninvasive RNA sampling to follow the early proinflammatory response of MEC during the course of a bacterial infection and showed that the levels of mRNA encoding SAA3 sharply increased at 24h postinfection. Taken together, our results demonstrate that MFG represent a unique source of MEC RNA to noninvasively sample sufficient amounts of high-quality RNA to assess the dynamics of MEC gene expression in vivo, especially during the first steps of infection, thereby paving the way for the discovery of early biomarkers for the control of intramammary infections. Furthermore, this noninvasive technique could be used to provide mammary transcriptomic data on a large scale, thus filling the gap between genomic and phenotypic data.
Collapse
Affiliation(s)
- P Brenaut
- INRA, UMR1313 Unité Génétique Animale et Biologie Intégrative, équipe «Lait, Génome & Santé» F-78350 Jouy-en-Josas, France
| | | | | | | | | | | |
Collapse
|
73
|
Oliver CH, Khaled WT, Frend H, Nichols J, Watson CJ. The Stat6-regulated KRAB domain zinc finger protein Zfp157 regulates the balance of lineages in mammary glands and compensates for loss of Gata-3. Genes Dev 2012; 26:1086-97. [PMID: 22588720 DOI: 10.1101/gad.184051.111] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Lineage commitment studies in mammary glands have focused on identifying cell populations that display stem or progenitor properties. However, the mechanisms that control cell fate have been incompletely explored. Herein we show that zinc finger protein 157 (Zfp157) is required to establish the balance between luminal alveolar pStat5- and Gata-3-expressing cells in the murine mammary gland. Using mice in which the zfp157 gene was disrupted, we found that alveologenesis was accelerated concomitantly with a dramatic skewing of the proportion of pStat5-expressing cells relative to Gata-3⁺ cells. This suppression of the Gata-3⁺ lineage was associated with increased expression of the inhibitor of helix-loop-helix protein Id2. Surprisingly, Gata-3 becomes dispensable in the absence of Zfp157, as mice deficient for both Zfp157 and Gata-3 lactate normally, although the glands display a mild epithelial dysplasia. These data suggest that the luminal alveolar compartment of the mammary gland is comprised of a number of distinct cell populations that, although interdependant, exhibit considerable cell fate plasticity.
Collapse
Affiliation(s)
- Carrie H Oliver
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom
| | | | | | | | | |
Collapse
|
74
|
Hughes K, Wickenden JA, Allen JE, Watson CJ. Conditional deletion of Stat3 in mammary epithelium impairs the acute phase response and modulates immune cell numbers during post-lactational regression. J Pathol 2012; 227:106-17. [PMID: 22081431 PMCID: PMC3477635 DOI: 10.1002/path.3961] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 10/04/2011] [Accepted: 10/31/2011] [Indexed: 12/31/2022]
Abstract
Mammary gland regression following weaning (involution) is associated with extensive cell death and the acquisition of an inflammatory signature. Characterizing the interplay between mammary epithelial cells, the re-emerging stroma and immune cells has implications for the understanding of the pathogenesis of pregnancy-associated breast cancer. Stat3 has a role in orchestrating cell death and involution, and we sought to determine whether expression of Stat3 by the mammary epithelium also influences the innate immune environment and inflammatory cell influx in the gland. We examined mice in which Stat3 is conditionally deleted only in the mammary epithelium. Distinct sets of genes associated with the acute phase response and innate immunity are markedly up-regulated during first phase involution in a Stat3-dependent manner. During second phase involution, chitinase 3-like 1, which has been associated with wound healing and chronic inflammatory conditions, is dramatically up-regulated by Stat3. Also at this time, the number of mammary macrophages and mast cells increases per unit area, and this increase is impaired in the absence of epithelial Stat3. Furthermore, expression of arginase-1 and Ym1, markers of alternatively activated macrophages, is significantly decreased in the absence of Stat3, whilst iNOS, a marker associated with classically activated macrophages, shows significantly increased expression in the Stat3-deleted glands. Thus, Stat3 is a key transcriptional regulator of genes associated with innate immunity and wound healing and influences mammary macrophage and mast cell numbers. The presence of epithelial Stat3 appears to polarize the macrophages and epithelial cells towards an alternatively activated phenotype, since in the absence of Stat3, the gland retains a phenotype associated with classically activated macrophages. These findings have relevance to the study of pregnancy-associated breast cancer and the role of Stat3 signalling in recruitment of alternatively activated tumour-associated macrophages in breast cancer. Copyright © 2012 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
|
75
|
Wagner KU, Schmidt JW. The two faces of Janus kinases and their respective STATs in mammary gland development and cancer. J Carcinog 2011; 10:32. [PMID: 22279417 PMCID: PMC3262999 DOI: 10.4103/1477-3163.90677] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 10/07/2011] [Indexed: 01/07/2023] Open
Abstract
Since its discovery as “just another kinase” more than twenty years ago, the family of JAK tyrosine kinases and their respective Signal Transducers and Activators of Transcription (STATs) has been a center of attention in the areas of signal transduction, development, and cancer. The subsequent designation of JAKs as Janus kinases after the mythical two-faced Roman God of the doorways accurately portrays the analogous and sometimes contrasting molecular and biological characteristics of these tyrosine kinases. The two “faces” of JAKs are their structurally similar kinase and pseudo-kinase domains. As essential parts of various transmembrane receptor complexes, these tyrosine kinases function at cellular gateways and relay signals from growth factors to their respective intracellular targets. The multifaceted nature of JAKs becomes evident from their ability to activate specific STATs during distinct phases of normal mammary gland development. Studies in breast cancer cells and genetically engineered mouse models also show that JAK/STAT signaling possesses a "two-faced" role during breast cancer initiation and progression. This review will highlight recent findings about important biological functions of JAKs and STATs during normal mammogenesis, with particular emphasis on the Jak2/Stat5 pathway as well as Jak1/2/Stat3 signaling complexes. In addition, we will discuss how the importance of these signaling networks changes during carcinogenesis. With JAK inhibitors currently under development to treat myeloproliferative disorders, determining the essential functions of JAKs at particular stages of disease initiation and progression is of critical importance to predict the efficacy of these agents for targeted therapies against breast cancer.
Collapse
Affiliation(s)
- Kay-Uwe Wagner
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, DRC2, Rm. 5033, Omaha, NE, USA
| | | |
Collapse
|
76
|
Wang Y, Li X, Hu H. Transcriptional regulation of co-expressed microRNA target genes. Genomics 2011; 98:445-52. [PMID: 22002038 DOI: 10.1016/j.ygeno.2011.09.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 08/12/2011] [Accepted: 09/24/2011] [Indexed: 01/26/2023]
Abstract
MicroRNAs play pivotal roles in gene regulation. Despite various research efforts on microRNAs, how microRNA target genes are transcriptionally regulated and how the transcriptional regulation of microRNA target genes relates to that of the microRNA genes are not well studied. By investigating the transcriptional regulation of microRNA target genes, we found that different groups of target genes of the same microRNA are co-expressed under different conditions, and these groups rarely overlap with each other for the majority of microRNAs. We also discovered that co-expressed microRNA target genes are often co-regulated, and different groups of target genes of the same microRNA are often regulated differently. In addition, we observed that transcription factors regulating a microRNA gene often regulate its target genes. Our study sheds light on the regulation of microRNA target genes, which will facilitate the prediction of microRNA target genes and the understanding of the transcriptional regulation of microRNA genes.
Collapse
Affiliation(s)
- Ying Wang
- Department of Electrical Engineering and Computer Science, University of Central Florida, Orlando, FL 32816, USA
| | | | | |
Collapse
|
77
|
Campbell JJ, Davidenko N, Caffarel MM, Cameron RE, Watson CJ. A multifunctional 3D co-culture system for studies of mammary tissue morphogenesis and stem cell biology. PLoS One 2011; 6:e25661. [PMID: 21984937 PMCID: PMC3184152 DOI: 10.1371/journal.pone.0025661] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 09/08/2011] [Indexed: 12/21/2022] Open
Abstract
Studies on the stem cell niche and the efficacy of cancer therapeutics require complex multicellular structures and interactions between different cell types and extracellular matrix (ECM) in three dimensional (3D) space. We have engineered a 3D in vitro model of mammary gland that encompasses a defined, porous collagen/hyaluronic acid (HA) scaffold forming a physiologically relevant foundation for epithelial and adipocyte co-culture. Polarized ductal and acinar structures form within this scaffold recapitulating normal tissue morphology in the absence of reconstituted basement membrane (rBM) hydrogel. Furthermore, organoid developmental outcome can be controlled by the ratio of collagen to HA, with a higher HA concentration favouring acinar morphological development. Importantly, this culture system recapitulates the stem cell niche as primary mammary stem cells form complex organoids, emphasising the utility of this approach for developmental and tumorigenic studies using genetically altered animals or human biopsy material, and for screening cancer therapeutics for personalised medicine.
Collapse
Affiliation(s)
- Jonathan J. Campbell
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
- * E-mail: (JJC); (CJW)
| | - Natalia Davidenko
- Department of Materials Science and Metallurgy, University of Cambridge, Cambridge, United Kingdom
| | - Maria M. Caffarel
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Ruth E. Cameron
- Department of Materials Science and Metallurgy, University of Cambridge, Cambridge, United Kingdom
| | - Christine J. Watson
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
- * E-mail: (JJC); (CJW)
| |
Collapse
|
78
|
Caffarel MM, Zaragoza R, Pensa S, Li J, Green AR, Watson CJ. Constitutive activation of JAK2 in mammary epithelium elevates Stat5 signalling, promotes alveologenesis and resistance to cell death, and contributes to tumourigenesis. Cell Death Differ 2011; 19:511-22. [PMID: 21941370 DOI: 10.1038/cdd.2011.122] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Signalling through the janus kinase (JAK)/signal transducer and activator of transcription (Stat) pathway is required at different stages of mammary gland development, and this pathway is frequently hyper-activated in cancer, including tumours of the breast. Stats 3, 5 and 6 have important roles in the differentiation and survival of mammary alveolar cells, but somewhat paradoxically, both Stat3 and 5 can have oncogenic activity in the mammary gland. Constitutive activation of JAK2 could be anticipated to result in hyper-activation of Stats 1, 3, 5 and 6 with concomitant cell transformation, although the outcome is difficult to envisage, particularly since Stats 3 and 5 play opposing roles in normal mammary gland development. Here, we show that expression of a constitutively active JAK2 mutant, JAK2 V617F, leads to hyper-activation of Stat5 in mammary epithelial cells (MECs), and transgenic mice expressing JAK2 V617F specifically in the mammary gland exhibit accelerated alveologenesis during pregnancy and delayed post-lactational regression. Overexpressing JAK2 V617F in MECs in vitro results in elevated proliferation and resistance to cell death. Furthermore, constitutively active JAK2 enhances anchorage-independent cell growth in the presence of a co-operating oncogene and accelerates tumourigenesis in a xenograft model. Taken together, our results provide insights into signalling downstream of constitutively active JAK2 and could be important for understanding the molecular mechanisms of breast tumourigenesis.
Collapse
Affiliation(s)
- M M Caffarel
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, UK.
| | | | | | | | | | | |
Collapse
|
79
|
Abstract
Signal transducer and activator of transcription (STAT) proteins are critical mediators of cytokine signaling. Among the seven STAT proteins, STAT6 is activated by IL-4 and IL-13 and plays a predominant role in the immune system. However, there is increasing evidence that STAT6 may function in other tissues and organ systems. IL-4, IL-13, and STAT6 promote humoral immunity, clearance of helminthic parasites as well as the pathogenesis of allergic disorders like asthma, food allergies, and atopic dermatitis. In this review, we will describe our current understanding of the biological functions of STAT6 and summarize recent advances in understanding the molecular mechanisms by which STAT6 regulates transcription.
Collapse
Affiliation(s)
- Shreevrat Goenka
- HB Wells Center of Pediatric Research, Department of Pediatrics, Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, 46202, USA.
| | | |
Collapse
|
80
|
Wei J, Yee C, Ramanathan P, Bendall LJ, Williamson P. Variation in immunophenotype of lactating mice. J Reprod Immunol 2011; 89:178-84. [PMID: 21531466 DOI: 10.1016/j.jri.2011.02.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Revised: 02/01/2011] [Accepted: 02/03/2011] [Indexed: 11/30/2022]
Abstract
Immunological factors have been shown to play a crucial role in mammary remodelling in rodent models of lactation, particularly at the stage of mammary involution. However, the relationship between immunological factors and the ability of normal mammary gland to produce milk, as well as the genetic components contributing to lactation performance remain largely unknown. In this study, we assessed the lactation and immunological phenotypes of 11 inbred mouse strains, namely 129X1/SvJ (129), A/J, AKR, C3H/HeJ (C3H), CBA/CaH (CBA), C57BL/6J (C57), DBA/1J, DBA/2J, FVB/N (FVB), QSi5 and SJL/J (SJL) to identify potential links. Leukocyte analyses showed no direct link between the fraction of splenic leukocytes and lactation performance. However, significant strain differences were discovered in the fraction of CD8+ T lymphocytes (P=0.016) and CD11b+Gr-1 mid-low monocytes (P<0.001). Cytokine profiles in plasma were examined and a subset of plasma cytokines, namely CCL2, CCL3, CCL5, CSF2, CSF3, IL10, IL15, IL1B, IL4, IL5, IL7 and TNF, were fitted to a linear regression model for prediction of lactation performance (R-sq=62%, S=0.309). Significant strain differences in the plasma cytokine levels were also discovered amongst these inbred strains. Analysis of immunological phenotypes showed strong correlations between splenic immune cell subsets and their regulating cytokine levels in plasma. The results demonstrate the extent of genetic variability in the immunological phenotypes of lactating mice, and provide a basis for understanding the role of cytokines in milk production, and identifying potential biomarkers of lactation performance.
Collapse
Affiliation(s)
- Jerry Wei
- Faculty of Veterinary Science, The University of Sydney, Camperdown, NSW 2006, Australia.
| | | | | | | | | |
Collapse
|
81
|
Kreuzaler PA, Staniszewska AD, Li W, Omidvar N, Kedjouar B, Turkson J, Poli V, Flavell RA, Clarkson RWE, Watson CJ. Stat3 controls lysosomal-mediated cell death in vivo. Nat Cell Biol 2011; 13:303-9. [PMID: 21336304 DOI: 10.1038/ncb2171] [Citation(s) in RCA: 249] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Accepted: 12/15/2010] [Indexed: 01/05/2023]
Abstract
It is well established that lysosomes play an active role during the execution of cell death. A range of stimuli can lead to lysosomal membrane permeabilization (LMP), thus inducing programmed cell death without involvement of the classical apoptotic programme. However, these lysosomal pathways of cell death have mostly been described in vitro or under pathological conditions. Here we show that the physiological process of post-lactational regression of the mammary gland is accomplished through a non-classical, lysosomal-mediated pathway of cell death. We found that, during involution, lysosomes in the mammary epithelium undergo widespread LMP. Furthermore, although cell death through LMP is independent of executioner caspases 3, 6 and 7, it requires Stat3, which upregulates the expression of lysosomal proteases cathepsin B and L, while downregulating their endogenous inhibitor Spi2A (ref. 8). Our findings report a previously unknown, Stat3-regulated lysosomal-mediated pathway of cell death under physiological circumstances. We anticipate that these findings will be of major importance in the design of treatments for cancers such as breast, colon and liver, where cathepsins and Stat3 are commonly overexpressed and/or hyperactivated respectively.
Collapse
Affiliation(s)
- Peter A Kreuzaler
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Watson CJ, Oliver CH, Khaled WT. Cytokine signalling in mammary gland development. J Reprod Immunol 2011; 88:124-9. [PMID: 21255846 DOI: 10.1016/j.jri.2010.11.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 11/17/2010] [Accepted: 11/27/2010] [Indexed: 10/18/2022]
Abstract
Mammary gland development occurs in three distinct stages during the lifetime of the female mammal: in embryonic, pubertal and reproductive life. At each of these developmental stages, different signalling molecules induce changes in both the epithelium and the surrounding stroma. However, it is during pregnancy that the most dramatic changes occur, resulting in a massive increase in the number of epithelial cells and in their function. Pregnancy initiates the development of a new epithelial lineage, the alveolar cells, which form the milk-producing lobuloalveolar structures. These cells become redundant at the end of lactation and are removed in an exquisitely controlled process of tissue remodelling coupled with extensive cell death. All of these events require not only steroid hormones but also sequential signalling by cytokines. A recent surprising discovery was that the signalling pathways and cytokines that regulate lineage determination in T helper cells are also involved in mammary gland development during pregnancy.
Collapse
Affiliation(s)
- Christine J Watson
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK.
| | | | | |
Collapse
|
83
|
Coussens LM, Pollard JW. Leukocytes in mammary development and cancer. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a003285. [PMID: 21123394 DOI: 10.1101/cshperspect.a003285] [Citation(s) in RCA: 153] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Leukocytes, of both the innate and adaptive lineages, are normal cellular components of all tissues. These important cells not only are critical for regulating normal tissue homeostasis, but also are significant paracrine regulators of all physiologic and pathologic tissue repair processes. This article summarizes recent insights regarding the trophic roles of leukocytes at each stage of mammary gland development and during cancer development, with a focus on Murids and humans.
Collapse
Affiliation(s)
- Lisa M Coussens
- Department of Pathology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, 94143, USA
| | | |
Collapse
|
84
|
Pedroza-Gonzalez A, Xu K, Wu TC, Aspord C, Tindle S, Marches F, Gallegos M, Burton EC, Savino D, Hori T, Tanaka Y, Zurawski S, Zurawski G, Bover L, Liu YJ, Banchereau J, Palucka AK. Thymic stromal lymphopoietin fosters human breast tumor growth by promoting type 2 inflammation. ACTA ACUST UNITED AC 2011; 208:479-90. [PMID: 21339324 PMCID: PMC3058586 DOI: 10.1084/jem.20102131] [Citation(s) in RCA: 193] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The human breast tumor microenvironment can display features of T helper type 2 (Th2) inflammation, and Th2 inflammation can promote tumor development. However, the molecular and cellular mechanisms contributing to Th2 inflammation in breast tumors remain unclear. Here, we show that human breast cancer cells produce thymic stromal lymphopoietin (TSLP). Breast tumor supernatants, in a TSLP-dependent manner, induce expression of OX40L on dendritic cells (DCs). OX40L(+) DCs are found in primary breast tumor infiltrates. OX40L(+) DCs drive development of inflammatory Th2 cells producing interleukin-13 and tumor necrosis factor in vitro. Antibodies neutralizing TSLP or OX40L inhibit breast tumor growth and interleukin-13 production in a xenograft model. Thus, breast cancer cell-derived TSLP contributes to the inflammatory Th2 microenvironment conducive to breast tumor development by inducing OX40L expression on DCs.
Collapse
|
85
|
Chua ACL, Hodson LJ, Moldenhauer LM, Robertson SA, Ingman WV. Dual roles for macrophages in ovarian cycle-associated development and remodelling of the mammary gland epithelium. Development 2010; 137:4229-38. [PMID: 21068060 DOI: 10.1242/dev.059261] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Each ovarian cycle, the mammary gland epithelium rotates through a sequence of hormonally regulated cell proliferation, differentiation and apoptosis. These studies investigate the role of macrophages in this cellular turnover. Macrophage populations and their spatial distribution were found to fluctuate across the cycle. The number of macrophages was highest at diestrus, and the greatest number of macrophages in direct contact with epithelial cells occurred at proestrus. The physiological necessity of macrophages in mammary gland morphogenesis during the estrous cycle was demonstrated in Cd11b-Dtr transgenic mice. Ovariectomised mice were treated with estradiol and progesterone to stimulate alveolar development, and with the progesterone receptor antagonist mifepristone to induce regression of the newly formed alveolar buds. Macrophage depletion during alveolar development resulted in a reduction in both ductal epithelial cell proliferation and the number of alveolar buds. Macrophage depletion during alveolar regression resulted in an increased number of branch points and an accumulation of TUNEL-positive cells. These studies show that macrophages have two roles in the cellular turnover of epithelial cells in the cycling mammary gland; following ovulation, they promote the development of alveolar buds in preparation for possible pregnancy, and they remodel the tissue back to its basic architecture in preparation for a new estrous cycle.
Collapse
Affiliation(s)
- Angela C L Chua
- The Robinson Institute, Research Centre for Reproductive Health, and School of Paediatrics and Reproductive Health, University of Adelaide, Adelaide 5005, Australia
| | | | | | | | | |
Collapse
|
86
|
Abstract
Spatial patterning of cell behaviors establishes the regional differences within tissues that collectively develop branched organs into their characteristic treelike shapes. Here we show that the pattern of branching morphogenesis of three-dimensional (3D) engineered epithelial tissues is controlled in part by gradients of endogenous mechanical stress. We used microfabrication to build model mammary epithelial tissues of defined geometry that branched in a stereotyped pattern when induced with growth factors. Branches initiated from sites of high mechanical stress within the tissues, as predicted numerically and measured directly using 3D traction force microscopy. Branch sites were defined by activation of focal adhesion kinase (FAK), inhibition of which disrupted morphogenesis. Stress, FAK activation, and branching were all altered by manipulating cellular contractility, matrix stiffness, intercellular cohesion and tissue geometry. These data suggest that the pattern and magnitude of mechanical stress across epithelial tissues cooperate with biochemical signals to specify branching pattern.
Collapse
Affiliation(s)
- Nikolce Gjorevski
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544
| | - Celeste M. Nelson
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| |
Collapse
|
87
|
Reed JR, Schwertfeger KL. Immune cell location and function during post-natal mammary gland development. J Mammary Gland Biol Neoplasia 2010; 15:329-39. [PMID: 20730636 PMCID: PMC4204476 DOI: 10.1007/s10911-010-9188-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Accepted: 08/10/2010] [Indexed: 01/21/2023] Open
Abstract
Post-natal mammary gland development requires complex interactions between the epithelial cells and various cell types within the stroma. Recent studies have illustrated the importance of immune cells and their mediators during the various stages of mammary gland development. However, the mechanisms by which these immune cells functionally contribute to mammary gland development are only beginning to be understood. This review provides an overview of the localization of immune cells within the mammary gland during the various stages of post-natal mammary gland development. Furthermore, recent studies are summarized that illustrate the mechanisms by which these cells are recruited to the mammary gland and their functional roles in mammary gland development.
Collapse
|
88
|
Abstract
Normal development of the mammary gland proceeds via interactions between the epithelium and the mesenchyme that start during embryogenesis and continue during pubertal outgrowth and differentiation. The function of specific peptide growth factors that bind members of the receptor tyrosine kinase family and the cytokine receptor family are required at each stage. In many cases the peptides are produced in one compartment and act on receptors in the other compartment. One of the striking differences between normal development and cancer is the loss of this cross-talk. Mammary tumor cells often produce a peptide and express the receptor on the same cell leading to autocrine activation of signaling pathways, a mechanism that is characteristic for cancer cells. We will discuss different peptides in the context of normal development and cancer in this review.
Collapse
Affiliation(s)
- Nancy E Hynes
- Friedrich Miescher Institute for Biomedical Research, Maulbeerestrasse 66, CH-4058 Basel, Switzerland.
| | | |
Collapse
|
89
|
Kiesler P, Haynes PA, Shi L, Kao PN, Wysocki VH, Vercelli D. NF45 and NF90 regulate HS4-dependent interleukin-13 transcription in T cells. J Biol Chem 2010; 285:8256-67. [PMID: 20051514 PMCID: PMC2832977 DOI: 10.1074/jbc.m109.041004] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Revised: 12/10/2009] [Indexed: 01/13/2023] Open
Abstract
Expression of the cytokine interleukin-13 (IL13) is critical for Th2 immune responses and Th2-mediated allergic diseases. Activation of human IL13 expression involves chromatin remodeling and formation of multiple DNase I-hypersensitive sites throughout the locus. Among these, HS4 is detected in the distal IL13 promoter in both naive and polarized CD4(+) T cells. We show herein that HS4 acts as a position-independent, orientation-dependent positive regulator of IL13 proximal promoter activity in transiently transfected, activated human CD4(+) Jurkat T cells and primary murine Th2 cells. The 3'-half of HS4 (HS4-3') was responsible for IL13 up-regulation and bound nuclear factor (NF) 90 and NF45, as demonstrated by DNA affinity chromatography coupled with tandem mass spectrometry, chromatin immunoprecipitation, and gel shift analysis. Notably, the CTGTT NF45/NF90-binding motif within HS4-3' was critical for HS4-dependent up-regulation of IL13 expression. Moreover, transfection of HS4-IL13 reporter vectors into primary, in vitro differentiated Th2 cells from wild-type, NF45(+/-), or NF90(+/-) mice showed that HS4 activity was exquisitely dependent on the levels of endogenous NF45 (and to a lesser degree NF90), because HS4-dependent IL13 expression was virtually abrogated in NF45(+/-) cells and reduced in NF90(+/-) cells. Collectively, our results identify NF45 and NF90 as novel regulators of HS4-dependent human IL13 transcription in response to T cell activation.
Collapse
Affiliation(s)
- Patricia Kiesler
- From the Functional Genomics Laboratory, Arizona Respiratory Center
| | | | - Lingfang Shi
- the Department of Pulmonary and Critical Care Medicine, Stanford University Medical Center, Stanford, California 94305
| | - Peter N. Kao
- the Department of Pulmonary and Critical Care Medicine, Stanford University Medical Center, Stanford, California 94305
| | | | - Donata Vercelli
- From the Functional Genomics Laboratory, Arizona Respiratory Center
- Cell Biology
- Arizona Center for the Biology of Complex Diseases, and
- The Bio5 Institute, University of Arizona, Tucson, Arizona 85719 and
| |
Collapse
|
90
|
Avril-Sassen S, Goldstein LD, Stingl J, Blenkiron C, Le Quesne J, Spiteri I, Karagavriilidou K, Watson CJ, Tavaré S, Miska EA, Caldas C. Characterisation of microRNA expression in post-natal mouse mammary gland development. BMC Genomics 2009; 10:548. [PMID: 19930549 PMCID: PMC2784809 DOI: 10.1186/1471-2164-10-548] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Accepted: 11/20/2009] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The differential expression pattern of microRNAs (miRNAs) during mammary gland development might provide insights into their role in regulating the homeostasis of the mammary epithelium. Our aim was to analyse these regulatory functions by deriving a comprehensive tissue-specific combined miRNA and mRNA expression profile of post-natal mouse mammary gland development.We measured the expression of 318 individual murine miRNAs by bead-based flow-cytometric profiling of whole mouse mammary glands throughout a 16-point developmental time course, including juvenile, puberty, mature virgin, gestation, lactation, and involution stages. In parallel whole-genome mRNA expression data were obtained. RESULTS One third (n = 102) of all murine miRNAs analysed were detected during mammary gland development. MicroRNAs were represented in seven temporally co-expressed clusters, which were enriched for both miRNAs belonging to the same family and breast cancer-associated miRNAs. Global miRNA and mRNA expression was significantly reduced during lactation and the early stages of involution after weaning. For most detected miRNA families we did not observe systematic changes in the expression of predicted targets. For miRNA families whose targets did show changes, we observed inverse patterns of miRNA and target expression. The data sets are made publicly available and the combined expression profiles represent an important community resource for mammary gland biology research. CONCLUSION MicroRNAs were expressed in likely co-regulated clusters during mammary gland development. Breast cancer-associated miRNAs were significantly enriched in these clusters. The mechanism and functional consequences of this miRNA co-regulation provide new avenues for research into mammary gland biology and generate candidates for functional validation.
Collapse
Affiliation(s)
- Stefanie Avril-Sassen
- Breast Cancer Functional Genomics Laboratory, Cancer Research UK Cambridge Research Institute and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Cambridge, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Faucon F, Rebours E, Bevilacqua C, Helbling JC, Aubert J, Makhzami S, Dhorne-Pollet S, Robin S, Martin P. Terminal differentiation of goat mammary tissue during pregnancy requires the expression of genes involved in immune functions. Physiol Genomics 2009; 40:61-82. [PMID: 19843654 DOI: 10.1152/physiolgenomics.00032.2009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Terminal differentiation of mammary tissue into a functional epithelium that synthesizes and secretes milk occurs during pregnancy. The molecular mechanisms underlying this complex process are poorly understood, especially in ruminants. To obtain an overview of the ruminant mammary gland's final differentiation process, we conducted time-course gene expression analysis of five physiological stages: four during pregnancy (P46, P70, P90, and P110) and one after 40 days of lactation (L40). An appropriate loop experimental design was used to follow gene expression profiles. Using three nulliparous (pregnancy) or primiparous (lactation) goats per stage, we performed a comparison starting from nine dye-swaps and using a 22K bovine oligoarray. Statistical analysis revealed that the expression of 1,696 genes varied significantly at least once in the study. These genes fell into 19 clusters based on their expression profiles. Identification of biological functions with Ingenuity Pathway Analysis software revealed several similarities, in keeping with physiological stages described in mice. As in mice, expression of milk protein genes began at midpregnancy, and genes regulating lipid biosynthesis were induced at the onset of lactation. During the first half of pregnancy, the molecular signature of goat mammary tissue was characterized by the expression of genes associated with tissue remodeling and differentiation, while the second half was mainly characterized by the presence of messengers encoding genes involved in cell proliferation. A large number of immune-related genes were also induced, supporting recent speculation that mammary tissue has an original immune function, and the recruitment of migrating hematopoietic cells possibly involved in the branching morphogenesis of the mammary gland. These data hint that the induction of differentiation occurs early in pregnancy, very likely before P46. This period is therefore crucial for obtaining a healthy and productive mammary gland.
Collapse
Affiliation(s)
- F Faucon
- Institut National de la Recherche Agronomique (INRA), Unité Mixte de Recherche (UMR) 1313, Génétique animale et Biologie intégrative, équipe LGS, F-78352 Jouy-en-Josas, France
| | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Cancer stem cells in breast cancer and metastasis. Breast Cancer Res Treat 2009; 118:241-54. [DOI: 10.1007/s10549-009-0524-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2009] [Accepted: 08/22/2009] [Indexed: 12/21/2022]
|
93
|
|
94
|
Reed JR, Leon RP, Hall MK, Schwertfeger KL. Interleukin-1beta and fibroblast growth factor receptor 1 cooperate to induce cyclooxygenase-2 during early mammary tumourigenesis. Breast Cancer Res 2009; 11:R21. [PMID: 19393083 PMCID: PMC2688950 DOI: 10.1186/bcr2246] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Revised: 03/17/2009] [Accepted: 04/24/2009] [Indexed: 12/14/2022] Open
Abstract
Introduction Inflammation within the tumour microenvironment correlates with increased invasiveness and poor prognosis in many types of cancer, including breast cancer. We have previously demonstrated that activation of a mouse mammary tumour virus (MMTV)-driven inducible fibroblast growth factor receptor 1 (iFGFR1) transgene in mammary epithelial cells results in an inflammatory response characterised by induction of inflammatory genes in the mammary gland. Specifically, we have observed increased levels of IL-1β expression in the mammary gland following activation of iFGFR1 and have used the iFGFR1 model to elucidate the function of IL-1β in promoting iFGFR1-induced mammary lesions. Methods To determine the functional consequences of IL-1β induction during FGFR1-induced mammary tumourigenesis, the effects of IL-1β inhibition on the formation of epithelial hyperplasias were examined using the MMTV-iFGFR1 transgenic mouse model. Further studies used a combination of the HC-11 mammary epithelial cell line that stably expresses iFGFR1 and the MMTV-iFGFR1 transgenic mice to further define the mechanisms of IL-1β function. Results Inhibition of IL-1β activity in vivo resulted in reduced iFGFR1-induced epithelial proliferation and formation of hyperplastic structures. Further studies demonstrated that treatment of mammary epithelial cells with IL-1β-induced expression of cyclooxygenase (Cox)-2 both in vitro and in vivo. Finally, inhibition of Cox-2 prior to activation of iFGFR1 in the transgenic mice also resulted in decreased iFGFR1-induced formation of hyperplastic structures. Conclusions The results from these studies indicate that targeting the inflammatory cytokine IL-1β partially inhibits iFGFR1-induced formation of early-stage mammary lesions, in part through induction of Cox-2. These findings demonstrate that activation of a growth factor receptor in mammary epithelial cells results in increased expression of inflammatory mediators, which cooperate to promote the initiation of hyperplastic lesions in the mammary gland.
Collapse
Affiliation(s)
- Johanna R Reed
- Microbiology, Immunology and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
95
|
Capaldo CT, Nusrat A. Cytokine regulation of tight junctions. BIOCHIMICA ET BIOPHYSICA ACTA 2009; 1788:864-71. [PMID: 18952050 PMCID: PMC2699410 DOI: 10.1016/j.bbamem.2008.08.027] [Citation(s) in RCA: 553] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/12/2008] [Revised: 08/26/2008] [Accepted: 08/27/2008] [Indexed: 01/11/2023]
Abstract
Epithelial and endothelial tight junctions act as a rate-limiting barrier between an organism and its environment. Continuing studies have highlighted the regulation of the tight junction barrier by cytokines. Elucidation of this interplay is vital for both the understanding of physiological tight junction regulation and the etiology of pathological conditions. This review will focus on recent advances in our understanding of the molecular mechanisms of tight junctions modulation by cytokines.
Collapse
Affiliation(s)
- Christopher T Capaldo
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA
| | | |
Collapse
|
96
|
Pérez-López FR, Chedraui P, Haya J. Review article: vitamin D acquisition and breast cancer risk. Reprod Sci 2009; 16:7-19. [PMID: 19144887 DOI: 10.1177/1933719108327595] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE The aim of the study was to focus on the association of vitamin D and breast cancer. METHODS The study of evidence concerning vitamin D's influence on the origin and development of breast cancer from a PubMed and individual searches. RESULTS Body sunlight exposure may reduce the prevalence of breast cancer. However, these studies correspond to global populations of different countries and regions without considering other geographic factors and individual, ethnic, and cultural factors that may affect sunlight exposure. Epidemiological analyses show that low vitamin D ingestion is associated with increased risk of breast cancer. Studies measuring serum vitamin D metabolites in women who were followed many years suggest that low circulating 25-hydroxyvitamin D3 levels are associated with increased breast cancer risk. CONCLUSIONS Although there are controversial results, it seems plausible that sufficient endogenous vitamin D levels may have a protective function on mammary cells, reducing breast cancer risk.
Collapse
Affiliation(s)
- Faustino R Pérez-López
- Department of Obstetrics and Gynecology, University of Zaragoza Faculty of Medicine, Clínico de Zaragoza Hospital, Domingo Miral s/n, Zaragoza, Spain.
| | | | | |
Collapse
|
97
|
Li W, Ferguson BJ, Khaled WT, Tevendale M, Stingl J, Poli V, Rich T, Salomoni P, Watson CJ. PML depletion disrupts normal mammary gland development and skews the composition of the mammary luminal cell progenitor pool. Proc Natl Acad Sci U S A 2009; 106:4725-30. [PMID: 19261859 PMCID: PMC2660741 DOI: 10.1073/pnas.0807640106] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Indexed: 01/08/2023] Open
Abstract
Nuclear domains of promyelocytic leukemia protein (PML) are known to act as signaling nodes in many cellular processes. Although the impact of PML expression in driving cell fate decisions for injured cells is well established, the function of PML in the context of tissue development is less well understood. Here, the in vivo role of PML in developmental processes in the murine mammary gland has been investigated. Data are presented showing that expression of PML is tightly regulated by three members of the Stat family of transcription factors that orchestrate the functional development of the mammary secretory epithelium during pregnancy. Developmental phenotypes were also discovered in the virgin and pregnant Pml null mouse, typified by aberrant differentiation of mammary epithelia with reduced ductal and alveolar development. PML depletion was also found to disturb the balance of two distinct luminal progenitor populations. Overall, it is shown that PML is required for cell lineage determination in bi-potent luminal progenitor cells and that the precise regulation of PML expression is required for functional differentiation of alveolar cells.
Collapse
Affiliation(s)
- Wenjing Li
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - Brian J. Ferguson
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - Walid T. Khaled
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - Maxine Tevendale
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - John Stingl
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - Valeria Poli
- Molecular Biotechnology Center, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Tina Rich
- Faculty of Veterinary Medicine, Institute of Comparative Medicine, 464 Bearsden Road, Glasgow G61 1QH, United Kingdom; and
| | - Paolo Salomoni
- MRC Toxicology Unit, Lancaster Road, P.O. Box 138, Leicester LE1 9HN, United Kingdom
| | - Christine J. Watson
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| |
Collapse
|
98
|
Abstract
Dramatic changes in cell composition and function occur in the mammary gland during a pregnancy-lactation-involution cycle. We investigated the transcriptional changes associated with these biological events by using microarray analysis and identified the critical genes involved by using genetically modified mice. Two surprising findings arose from these studies. First, the microarray data showed that postlactational regression was associated with an acute phase inflammatory response, in addition to cell death. Conditional deletion of signal transducer and activator of transcription (Stat)3 or the nuclear factor-kappaB regulatory kinase inhibitor of kappa B kinase beta resulted in a failure of cell death induction during involution, an indication that these signaling pathways are essential mediators of the involution process. Both Stat3 and nuclear factor-kappaB have been shown to regulate acute phase gene expression in addition to apoptosis regulators. Four distinct transcriptional profiles are present in the first 4 d of involution, whereas there are 3 in lactation. At the peak of lactation (i.e., d 10 in mouse), more than 400 genes reach their maximum expression before declining dramatically in the first 12 h of involution. A reciprocal pattern was observed for more than 500 genes that were specifically upregulated within the first 12 h of forced involution. We are now investigating the role of a subset of these genes in involution. We also uncovered a role for genes normally associated with immune cell signaling in the differentiation of luminal mammary epithelial cells during pregnancy. Genetic deletion of the transcription factor Stat6 resulted in delayed development during pregnancy, and this phenotype was recapitulated in mammary tissue from IL-4 and IL-13 doubly deficient mice. Furthermore, we showed that mammary epithelial cells secrete T-cell regulatory cytokines. T-helper type 1 cytokines, such as interferon-gamma and IL-12a, are secreted by undifferentiated mammary epithelial cells, whereas T-helper type 2 cytokines, including IL-4 and IL-13, are secreted by differentiated cells. This unexpected finding demonstrates a role for immune cell signaling in mammary epithelial cell fate and function.
Collapse
Affiliation(s)
- C J Watson
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom.
| |
Collapse
|
99
|
Watson CJ, Neoh K. The Stat family of transcription factors have diverse roles in mammary gland development. Semin Cell Dev Biol 2008; 19:401-6. [PMID: 18723104 DOI: 10.1016/j.semcdb.2008.07.021] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Revised: 07/22/2008] [Accepted: 07/24/2008] [Indexed: 10/21/2022]
Abstract
The Stat family of transcription factors have diverse roles in mammary gland development. Genetic studies in mice have revealed an essential requirement for Stat5a in development of secretory alveolar cells during pregnancy while Stat6, which is normally associated with differentiation of T helper cells, is important in the commitment of luminal cells to this alveolar lineage. In contrast, Stat3 is specifically activated at the initiation of post-lactational regression when it has an essential function in the regulation of cell death and tissue remodelling. Stat1 and Stat4 have been shown to be regulated during a mammary developmental cycle although whether they have specific, non-redundant roles is not clear. Thus, the adult mammary gland is somewhat unusual in that it is a tissue where different Stats are sequentially activated to orchestrate the processes of functional differentiation, cell death and tissue remodelling.
Collapse
Affiliation(s)
- C J Watson
- University of Cambridge, Department of Pathology, Tennis Court Road, Cambridge CB2 1QP, United Kingdom.
| | | |
Collapse
|
100
|
Disclosing JAK/STAT links to cell adhesion and cell polarity. Semin Cell Dev Biol 2008; 19:370-8. [DOI: 10.1016/j.semcdb.2008.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Revised: 06/05/2008] [Accepted: 06/06/2008] [Indexed: 12/27/2022]
|