101
|
Abstract
Human cancers exhibit formidable molecular heterogeneity, to a large extent accounting for the incomplete and transitory efficacy of current anti-cancer therapies. However, neoplastic cells alone do not manifest the disease, but conscript a battery of non-tumor cells to enable and sustain hallmark capabilities of cancer. Escaping immunosurveillance is one of such capabilities. Tumors evolve immunosuppressive microenvironment to subvert anti-tumor immunity. In this review, we will focus on tumor-associated myeloid cells, which constitute an essential part of the immune microenvironment and reciprocally interact with cancer cells to establish malignancy toward metastasis. The diversity and plasticity of these cells constitute another layer of heterogeneity, beyond the heterogeneity of cancer cells themselves. We envision that immune microenvironment co-evolves with the genetic heterogeneity of tumor. Addressing the question of how genetically distinct tumors shape and are shaped by unique immune microenvironment will provide an attractive rationale to develop novel immunotherapeutic modalities. Here, we discuss the complex nature of tumor microenvironment, with an emphasis on the cellular and functional heterogeneity among tumor-associated myeloid cells as well as immune environment heterogeneity in the context of a full spectrum of human breast cancers.
Collapse
|
102
|
Sattler S. The Role of the Immune System Beyond the Fight Against Infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1003:3-14. [PMID: 28667551 DOI: 10.1007/978-3-319-57613-8_1] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The immune system was identified as a protective factor during infectious diseases over a century ago. Current definitions and textbook information are still largely influenced by these early observations, and the immune system is commonly presented as a defence machinery. However, host defence is only one manifestation of the immune system's overall function in the maintenance of tissue homeostasis and system integrity. In fact, the immune system is integral part of fundamental physiological processes such as development, reproduction and wound healing, and a close crosstalk between the immune system and other body systems such as metabolism, the central nervous system and the cardiovascular system is evident. Research and medical professionals in an expanding range of areas start to recognise the implications of the immune system in their respective fields.This chapter provides a brief historical perspective on how our understanding of the immune system has evolved from a defence system to an overarching surveillance machinery to maintain tissue integrity. Current perspectives on the non-defence functions of classical immune cells and factors will also be discussed.
Collapse
Affiliation(s)
- Susanne Sattler
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
103
|
Munro DAD, Hughes J. The Origins and Functions of Tissue-Resident Macrophages in Kidney Development. Front Physiol 2017; 8:837. [PMID: 29118719 PMCID: PMC5660965 DOI: 10.3389/fphys.2017.00837] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 10/09/2017] [Indexed: 12/21/2022] Open
Abstract
The adult kidney hosts tissue-resident macrophages that can cause, prevent, and/or repair renal damage. Most of these macrophages derive from embryonic progenitors that colonize the kidney during its development and proliferate in situ throughout adulthood. Although the precise origins of kidney macrophages remain controversial, recent studies have revealed that embryonic macrophage progenitors initially migrate from the yolk sac, and later from the fetal liver, into the developing kidney. Once in the kidney, tissue-specific transcriptional regulators specify macrophage progenitors into dedicated kidney macrophages. Studies suggest that kidney macrophages facilitate many processes during renal organogenesis, such as branching morphogenesis and the clearance of cellular debris; however, little is known about how the origins and specification of kidney macrophages dictate their function. Here, we review significant new findings about the origins, specification, and developmental functions of kidney macrophages.
Collapse
Affiliation(s)
- David A D Munro
- Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Jeremy Hughes
- MRC Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
104
|
Berryhill GE, Lemay DG, Trott JF, Aimo L, Lock AL, Hovey RC. The Transcriptome of Estrogen-Independent Mammary Growth in Female Mice Reveals That Not All Mammary Glands Are Created Equally. Endocrinology 2017; 158:3126-3139. [PMID: 28938404 PMCID: PMC5659702 DOI: 10.1210/en.2017-00395] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Accepted: 07/21/2017] [Indexed: 01/22/2023]
Abstract
Allometric growth of ducts in the mammary glands (MGs) is widely held to be estrogen dependent. We previously discovered that the dietary fatty acid trans-10, cis-12 conjugated linoleic acid (CLA) stimulates estrogen-independent allometric growth and terminal end bud formation in ovariectomized mice. Given the similar phenotype induced by estrogen and CLA, we investigated the shared and/or divergent mechanisms underlying these changes. We confirmed MG growth induced by CLA is temporally distinct from that elicited by estrogen. We then used RNA sequencing to compare the transcriptome of the MG during similar proliferative and morphological states. Both estrogen and CLA affected the genes involved in proliferation. The transcriptome for estrogen-treated mice included canonical estrogen-induced genes, including Pgr, Areg, and Foxa1. In contrast, their expression was unchanged by CLA. However, CLA, but not estrogen, altered expression of a unique set of inflammation-associated genes, consistent with stromal changes. This CLA-altered signature included increased expression of epidermal growth factor receptor (EGFR) pathway components, consistent with the demonstration that CLA-induced MG growth is EGFR dependent. Our findings highlight a unique role for diet-induced inflammation that underlies estrogen-independent MG development.
Collapse
Affiliation(s)
- Grace E. Berryhill
- Department of Animal Science, University of California Davis, Davis, California 95616-8521
| | - Danielle G. Lemay
- UC Davis Genome Center, University of California Davis, Davis, California 95616-8521
- US Department of Agriculture, Agricultural Research Services, Western Human Nutrition Research Center, Davis, California 95616
| | - Josephine F. Trott
- Department of Animal Science, University of California Davis, Davis, California 95616-8521
| | - Lucila Aimo
- Department of Animal Science, University of California Davis, Davis, California 95616-8521
| | - Adam L. Lock
- Department of Animal Science, Michigan State University, East Lansing, Michigan 48824-1225
| | - Russell C. Hovey
- Department of Animal Science, University of California Davis, Davis, California 95616-8521
| |
Collapse
|
105
|
Tamma R, Guidolin D, Annese T, Tortorella C, Ruggieri S, Rega S, Zito FA, Nico B, Ribatti D. Spatial distribution of mast cells and macrophages around tumor glands in human breast ductal carcinoma. Exp Cell Res 2017; 359:179-184. [PMID: 28756894 DOI: 10.1016/j.yexcr.2017.07.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/17/2017] [Accepted: 07/26/2017] [Indexed: 01/06/2023]
Abstract
Macrophages and mast cells are usually present in the tumor microenvironment and play an important role as regulators of inflammation, immunological response and angiogenesis in the tumor microenvironment. In this study, we have evaluated macrophage, mast cell, and microvessel density in a selected group of different grade of invasive breast carcinoma tumor specimens. Furthermore, we have investigated the pattern of distribution of CD68-positive macrophages and tryptase-positive mast cells around tumor glands. Results have shown that: A) Macrophages are more numerous in G2 and G3 breast cancer stages respect to controls, the per cent of macrophages in G1 samples was comparable to the controls, and the spatial relationship between macrophages and glands (as indicated by the mean cell-to-gland distance) correlated with CD31-positive vessels. B) Mast cells in G2 and G3 tumor specimens show a significant increase in their number as compared to control samples, and their spatial distribution around the glands did not show any significant difference among groups. Overall, the results of this study confirm the important role of macrophages and mast cells in tumor progression and angiogenesis in human ductal breast cancer, and pointed out the spatial relationship between tumor macrophages and glands, and its correlation with microvascular density.
Collapse
Affiliation(s)
- Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; National Cancer Institute "Giovanni Paolo II", Bari, Italy
| | - Diego Guidolin
- Department of Neurosciences, University of Padova Medical School, University of Padova, Italy
| | - Tiziana Annese
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Cinzia Tortorella
- Department of Neurosciences, University of Padova Medical School, University of Padova, Italy
| | - Simona Ruggieri
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Serena Rega
- Department of Pathology "San Paolo" Hospital, Bari, Italy
| | | | - Beatrice Nico
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, Bari, Italy; National Cancer Institute "Giovanni Paolo II", Bari, Italy.
| |
Collapse
|
106
|
Marichal T, Mesnil C, Bureau F. Homeostatic Eosinophils: Characteristics and Functions. Front Med (Lausanne) 2017; 4:101. [PMID: 28744457 PMCID: PMC5504169 DOI: 10.3389/fmed.2017.00101] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/26/2017] [Indexed: 12/17/2022] Open
Abstract
Eosinophils are typically considered to be specialized effector cells that are recruited to the tissues as a result of T helper type 2 (Th2) cell responses associated with helminth infections or allergic diseases such as asthma. Once at the site of injury, eosinophils release their cytotoxic granule proteins as well as preformed cytokines and lipid mediators, contributing to parasite destruction but also to exacerbation of inflammation and tissue damage. Accumulating evidence indicates that, besides their roles in Th2 responses, eosinophils also regulate homeostatic processes at steady state, thereby challenging the exclusive paradigm of the eosinophil as a destructive and inflammatory cell. Indeed, under baseline conditions, eosinophils rapidly leave the bloodstream to enter tissues, mainly the gastrointestinal tract, lungs, adipose tissue, thymus, uterus, and mammary glands, where they regulate a variety of important biological functions, such as immunoregulation, control of glucose homeostasis, protection against obesity, regulation of mammary gland development, and preparation of the uterus for pregnancy. This article provides an overview of the characteristics and functions of these homeostatic eosinophils.
Collapse
Affiliation(s)
- Thomas Marichal
- Laboratory of Cellular and Molecular Immunology, GIGA-Research, University of Liège, Liège, Belgium.,Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Claire Mesnil
- Laboratory of Cellular and Molecular Immunology, GIGA-Research, University of Liège, Liège, Belgium.,Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Fabrice Bureau
- Laboratory of Cellular and Molecular Immunology, GIGA-Research, University of Liège, Liège, Belgium.,Faculty of Veterinary Medicine, University of Liège, Liège, Belgium.,WELBIO, Walloon Excellence in Life Sciences and Biotechnology, Wallonia, Belgium
| |
Collapse
|
107
|
STAT5 deletion in macrophages alters ductal elongation and branching during mammary gland development. Dev Biol 2017; 428:232-244. [PMID: 28606561 PMCID: PMC5621646 DOI: 10.1016/j.ydbio.2017.06.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/04/2017] [Accepted: 06/06/2017] [Indexed: 12/31/2022]
Abstract
Macrophages are required for proper mammary gland development and maintaining tissue homeostasis. However, the mechanisms by which macrophages regulate this process remain unclear. Here, we identify STAT5 as an important regulator of macrophage function in the developing mammary gland. Analysis of mammary glands from mice with STAT5-deficient macrophages demonstrates delayed ductal elongation, enhanced ductal branching and increased epithelial proliferation. Further analysis reveals that STAT5 deletion in macrophages leads to enhanced expression of proliferative factors such as Cyp19a1/aromatase and IL-6. Mechanistic studies demonstrate that STAT5 binds directly to the Cyp19a1 promoter in macrophages to suppress gene expression and that loss of STAT5 results in enhanced stromal expression of aromatase. Finally, we demonstrate that STAT5 deletion in macrophages cooperates with oncogenic initiation in mammary epithelium to accelerate the formation of estrogen receptor (ER)-positive hyperplasias. These studies establish the importance of STAT5 in macrophages during ductal morphogenesis in the mammary gland and demonstrate that altering STAT5 function in macrophages can affect the development of tissue-specific disease.
Collapse
|
108
|
Paine IS, Lewis MT. The Terminal End Bud: the Little Engine that Could. J Mammary Gland Biol Neoplasia 2017; 22:93-108. [PMID: 28168376 PMCID: PMC5488158 DOI: 10.1007/s10911-017-9372-0] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 01/17/2017] [Indexed: 12/12/2022] Open
Abstract
The mammary gland is one of the most regenerative organs in the body, with the majority of development occurring postnatally and in the adult mammal. Formation of the ductal tree is orchestrated by a specialized structure called the terminal end bud (TEB). The TEB is responsible for the production of mature cell types leading to the elongation of the subtending duct. The TEB is also the regulatory control point for basement membrane deposition, branching, angiogenesis, and pattern formation. While the hormonal control of TEB growth is well characterized, the local regulatory factors are less well understood. Recent studies of pubertal outgrowth and ductal elongation have yielded surprising details in regards to ongoing processes in the TEB. Here we summarize the current understanding of TEB biology, discuss areas of future study, and discuss the use of the TEB as a model for the study of breast cancer.
Collapse
Affiliation(s)
- Ingrid S Paine
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Michael T Lewis
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department Radiology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
109
|
Estrogen reprograms the activity of neutrophils to foster protumoral microenvironment during mammary involution. Sci Rep 2017; 7:46485. [PMID: 28429725 PMCID: PMC5399373 DOI: 10.1038/srep46485] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 03/21/2017] [Indexed: 12/14/2022] Open
Abstract
Epidemiological studies have indicated increased risk for breast cancer within 10 years of childbirth. Acute inflammation during mammary involution has been suggested to promote this parity-associated breast cancer. We report here that estrogen exacerbates mammary inflammation during involution. Microarray analysis shows that estrogen induces an extensive proinflammatory gene signature in the involuting mammary tissue. This is associated with estrogen-induced neutrophil infiltration. Furthermore, estrogen induces the expression of protumoral cytokines/chemokines, COX-2 and tissue-remodeling enzymes in isolated mammary neutrophils and systemic neutrophil depletion abolished estrogen-induced expression of these genes in mammary tissue. More interestingly, neutrophil depletion diminished estrogen-induced growth of ERα-negative mammary tumor 4T1 in Balb/c mice. These findings highlight a novel aspect of estrogen action that reprograms the activity of neutrophils to create a pro-tumoral microenvironment during mammary involution. This effect on the microenvironment would conceivably aggravate its known neoplastic effect on mammary epithelial cells.
Collapse
|
110
|
Law AMK, Lim E, Ormandy CJ, Gallego-Ortega D. The innate and adaptive infiltrating immune systems as targets for breast cancer immunotherapy. Endocr Relat Cancer 2017; 24:R123-R144. [PMID: 28193698 PMCID: PMC5425956 DOI: 10.1530/erc-16-0404] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 02/13/2017] [Indexed: 12/15/2022]
Abstract
A cancer cell-centric view has long dominated the field of cancer biology. Research efforts have focussed on aberrant cancer cell signalling pathways and on changes to cancer cell DNA. Mounting evidence demonstrates that many cancer-associated cell types within the tumour stroma co-evolve and support tumour growth and development, greatly modifying cancer cell behaviour, facilitating invasion and metastasis and controlling dormancy and sensitivity to drug therapy. Thus, these stromal cells represent potential targets for cancer therapy. Among these cell types, immune cells have emerged as a promising target for therapy. The adaptive and the innate immune system play an important role in normal mammary development and breast cancer. The number of infiltrating adaptive immune system cells with tumour-rejecting capacity, primarily, T lymphocytes, is lower in breast cancer compared with other cancer types, but infiltration occurs in a large proportion of cases. There is strong evidence demonstrating the importance of the immunosuppressive role of the innate immune system during breast cancer progression. A consideration of components of both the innate and the adaptive immune system is essential for the design and development of immunotherapies in breast cancer. In this review, we focus on the importance of immunosuppressive myeloid-derived suppressor cells (MDSCs) as potential targets for breast cancer therapy.
Collapse
Affiliation(s)
- Andrew M K Law
- Tumour Development GroupThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- Cancer Biology LaboratoryThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Elgene Lim
- Connie Johnson Breast Cancer Research LaboratoryThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical SchoolFaculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Christopher J Ormandy
- Cancer Biology LaboratoryThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical SchoolFaculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - David Gallego-Ortega
- Tumour Development GroupThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical SchoolFaculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia
| |
Collapse
|
111
|
Kimura M, Shimomura M, Morishita H, Meguro T, Seto S. Eosinophilia in infants with food protein-induced enterocolitis syndrome in Japan. Allergol Int 2017; 66:310-316. [PMID: 27624222 DOI: 10.1016/j.alit.2016.08.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 07/08/2016] [Accepted: 08/01/2016] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Many Japanese infants with food protein-induced enterocolitis syndrome (FPIES) show eosinophilia, which has been thought to be a characteristic of food protein-induced proctocolitis (FPIP). METHODS To elucidate the characteristics of eosinophilia in Japanese FPIES patients, 113 infants with non-IgE-mediated gastrointestinal food allergy due to cow's milk were enrolled and classified into FPIES (n = 94) and FPIP (n = 19). RESULTS The percentage of peripheral blood eosinophils (Eo) was increased in most FPIES patients (median, 7.5%), which was comparable with that in FPIP patients (9.0%). Among FPIES patients, Eo was the highest in patients who had vomiting, bloody stool, and diarrhea simultaneously (12.9%) and lowest in patients with diarrhea alone (3.2%). Eo showed a significant positive correlation with the incidence of vomiting (Cramer's V = 0.31, p < 0.005) and bloody stool (Cramer's V = 0.34, p < 0.0005). A significant difference was found in Eo between early- (≤10 days, n = 56) and late-onset (>10 days, n = 38) FPIES (median, 9.8% vs. 5.4%; p < 0.005). IL-5 production by peripheral blood T cells stimulated with cow's milk protein in early-onset FPIES was significantly higher than that in late-onset FPIES (67.7 pg/mL vs. 12.5 pg/mL, p < 0.01), and showed a significant positive correlation with Eo (rs = 0.60, p < 0.01). CONCLUSIONS This study demonstrated two types of eosinophilia in Japanese FPIES infants: conspicuous and mild eosinophilia in early- and late-onset FPIES patients, respectively. Conspicuous eosinophilia in early-onset FPIES is suggested to be caused by abnormally high IL-5 production.
Collapse
Affiliation(s)
- Mitsuaki Kimura
- Department of Allergy and Clinical Immunology, Shizuoka Children's Hospital, Shizuoka, Japan.
| | - Masaki Shimomura
- Department of Allergy and Clinical Immunology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Hideaki Morishita
- Department of Allergy and Clinical Immunology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Takaaki Meguro
- Department of Allergy and Clinical Immunology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Shiro Seto
- Department of Allergy and Clinical Immunology, Shizuoka Children's Hospital, Shizuoka, Japan
| |
Collapse
|
112
|
Zhao C, Cai S, Shin K, Lim A, Kalisky T, Lu WJ, Clarke MF, Beachy PA. Stromal Gli2 activity coordinates a niche signaling program for mammary epithelial stem cells. Science 2017; 356:science.aal3485. [PMID: 28280246 DOI: 10.1126/science.aal3485] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 03/01/2017] [Indexed: 12/14/2022]
Abstract
The stem cell niche is a complex local signaling microenvironment that sustains stem cell activity during organ maintenance and regeneration. The mammary gland niche must support its associated stem cells while also responding to systemic hormonal regulation that triggers pubertal changes. We find that Gli2, the major Hedgehog pathway transcriptional effector, acts within mouse mammary stromal cells to direct a hormone-responsive niche signaling program by activating expression of factors that regulate epithelial stem cells as well as receptors for the mammatrophic hormones estrogen and growth hormone. Whereas prior studies implicate stem cell defects in human disease, this work shows that niche dysfunction may also cause disease, with possible relevance for human disorders and in particular the breast growth pathogenesis associated with combined pituitary hormone deficiency.
Collapse
Affiliation(s)
- Chen Zhao
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shang Cai
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kunyoo Shin
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyumgbuk 37673, South Korea
| | - Agnes Lim
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tomer Kalisky
- Faculty of Engineering and Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 52900, Israel
| | - Wan-Jin Lu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael F Clarke
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Philip A Beachy
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA. .,Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA.,Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
113
|
Monkkonen T, Landua JD, Visbal AP, Lewis MT. Epithelial and non-epithelial Ptch1 play opposing roles to regulate proliferation and morphogenesis of the mouse mammary gland. Development 2017; 144:1317-1327. [PMID: 28275010 PMCID: PMC5399619 DOI: 10.1242/dev.140434] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 02/07/2017] [Indexed: 12/14/2022]
Abstract
Patched 1 (Ptch1) has epithelial, stromal and systemic roles in murine mammary gland organogenesis, yet specific functions remain undefined. Cre-recombinase-mediated Ptch1 ablation in mammary epithelium increased proliferation and branching, but did not phenocopy transgenic expression of activated smoothened (SmoM2). The epithelium showed no evidence of canonical hedgehog signaling, and hyperproliferation was not blocked by smoothened (SMO) inhibition, suggesting a non-canonical function of PTCH1. Consistent with this possibility, nuclear localization of cyclin B1 was increased. In non-epithelial cells, heterozygous Fsp-Cre-mediated Ptch1 ablation increased proliferation and branching, with dysplastic terminal end buds (TEB) and ducts. By contrast, homozygous Ptch1 ablation decreased proliferation and branching, producing stunted ducts filled with luminal cells showing altered ovarian hormone receptor expression. Whole-gland transplantation into wild-type hosts or estrogen/progesterone treatment rescued outgrowth and hormone receptor expression, but not the histological changes. Bone marrow transplantation failed to rescue outgrowth. Ducts of Fsp-Cre;Ptch1fl/fl mice were similar to Fsp-Cre;SmoM2 ducts, but Fsp-Cre;SmoM2 outgrowths were not stunted, suggesting that the histology might be mediated by Smo in the local stroma, with systemic Ptch1 required for ductal outgrowth and proper hormone receptor expression in the mammary epithelium. Summary: Systemic and tissue-specific depletion of patched 1 in epithelial and stromal compartments of the mammary gland defines functions in ductal patterning, proliferation and gene expression.
Collapse
Affiliation(s)
- Teresa Monkkonen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - John D Landua
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Adriana P Visbal
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.,Program in Developmental Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Michael T Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA .,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.,Program in Developmental Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
114
|
Dill R, Walker AM. Role of Prolactin in Promotion of Immune Cell Migration into the Mammary Gland. J Mammary Gland Biol Neoplasia 2017; 22:13-26. [PMID: 27900586 PMCID: PMC5313375 DOI: 10.1007/s10911-016-9369-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 11/21/2016] [Indexed: 01/13/2023] Open
Abstract
Immune cells in the mammary gland play a number of important roles, including protection against infection during lactation and, after passing into milk, modulation of offspring immunity. However, little is known about the mechanism of recruitment of immune cells to the lactating gland in the absence of infection. Given the importance of prolactin to other aspects of lactation, we hypothesized it would also play a role in immune cell recruitment. Prolactin treatment of adult female mice for a period equivalent to pregnancy and the first week of lactation increased immune cell flux through the mammary gland, as reflected in the number of immune cells in mammary gland-draining, but not other lymph nodes. Conditioned medium from luminal mammary epithelial HC11 cell cultures was chemo-attractive to CD4+ and CD8+ T cells, CD4+ and CD8+ memory T cells, B cells, macrophages, monocytes, eosinophils, and neutrophils. Prolactin did not act as a direct chemo-attractant, but through effects on luminal mammary epithelial cells, increased the chemo-attractant properties of conditioned medium. Macrophages and neutrophils constitute the largest proportion of cells in milk from healthy glands. Depletion of CCL2 and CXCL1 from conditioned medium reduced chemo-attraction of monocytes and neutrophils, and prolactin increased expression of these two chemokines in mammary epithelial cells. We conclude that prolactin is an important player in the recruitment of immune cells to the mammary gland both through its activities to increase epithelial cell number as well as production of chemo-attractants on a per cell basis.
Collapse
Affiliation(s)
- Riva Dill
- Division of Biomedical Sciences, School of Medicine, University of California, 900 University Ave. 1260 Webber Hall, Riverside, CA, 92521, USA.
| | - Ameae M Walker
- Division of Biomedical Sciences, School of Medicine, University of California, 900 University Ave. 1260 Webber Hall, Riverside, CA, 92521, USA
| |
Collapse
|
115
|
Carron EC, Homra S, Rosenberg J, Coffelt SB, Kittrell F, Zhang Y, Creighton CJ, Fuqua SA, Medina D, Machado HL. Macrophages promote the progression of premalignant mammary lesions to invasive cancer. Oncotarget 2017; 8:50731-50746. [PMID: 28881599 PMCID: PMC5584199 DOI: 10.18632/oncotarget.14913] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 12/26/2016] [Indexed: 12/17/2022] Open
Abstract
Breast cancer initiation, progression and metastasis rely on a complex interplay between tumor cells and their surrounding microenvironment. Infiltrating immune cells, including macrophages, promote mammary tumor progression and metastasis; however, less is known about the role of macrophages in early stage lesions. In this study, we utilized a transplantable p53-null model of early progression to characterize the immune cell components of early stage lesions. We show that macrophages are recruited to ductal hyperplasias with a high tumor-forming potential where they are differentiated and polarized toward a tumor-promoting phenotype. These macrophages are a unique subset of macrophages, characterized by pro-inflammatory, anti-inflammatory and immunosuppressive factors. Macrophage ablation studies showed that macrophages are required for both early stage progression and primary tumor formation. These studies suggest that therapeutic targeting of tumor-promoting macrophages may not only be an effective strategy to block tumor progression and metastasis, but may also have critical implications for breast cancer prevention.
Collapse
Affiliation(s)
- Emily C Carron
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, USA
| | - Samuel Homra
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, USA
| | - Jillian Rosenberg
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, USA
| | - Seth B Coffelt
- CRUK Beatson Institute and Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Frances Kittrell
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Yiqun Zhang
- Dan L. Duncan Comprehensive Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, TX, USA
| | - Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, TX, USA
| | - Suzanne A Fuqua
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Daniel Medina
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Heather L Machado
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, USA
| |
Collapse
|
116
|
Sun X, Glynn DJ, Hodson LJ, Huo C, Britt K, Thompson EW, Woolford L, Evdokiou A, Pollard JW, Robertson SA, Ingman WV. CCL2-driven inflammation increases mammary gland stromal density and cancer susceptibility in a transgenic mouse model. Breast Cancer Res 2017; 19:4. [PMID: 28077158 PMCID: PMC5225654 DOI: 10.1186/s13058-016-0796-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 12/07/2016] [Indexed: 12/16/2022] Open
Abstract
Background Macrophages play diverse roles in mammary gland development and breast cancer. CC-chemokine ligand 2 (CCL2) is an inflammatory cytokine that recruits macrophages to sites of injury. Although CCL2 has been detected in human and mouse mammary epithelium, its role in regulating mammary gland development and cancer risk has not been explored. Methods Transgenic mice were generated wherein CCL2 is driven by the mammary epithelial cell-specific mouse mammary tumour virus 206 (MMTV) promoter. Estrous cycles were tracked in adult transgenic and non-transgenic FVB mice, and mammary glands collected at the four different stages of the cycle. Dissected mammary glands were assessed for cyclical morphological changes, proliferation and apoptosis of epithelium, macrophage abundance and collagen deposition, and mRNA encoding matrix remodelling enzymes. Another cohort of control and transgenic mice received carcinogen 7,12-Dimethylbenz(a)anthracene (DMBA) and tumour development was monitored weekly. CCL2 protein was also quantified in paired samples of human breast tissue with high and low mammographic density. Results Overexpression of CCL2 in the mammary epithelium resulted in an increased number of macrophages, increased density of stroma and collagen and elevated mRNA encoding matrix remodelling enzymes lysyl oxidase (LOX) and tissue inhibitor of matrix metalloproteinases (TIMP)3 compared to non-transgenic controls. Transgenic mice also exhibited increased susceptibility to development of DMBA-induced mammary tumours. In a paired sample cohort of human breast tissue, abundance of epithelial-cell-associated CCL2 was higher in breast tissue of high mammographic density compared to tissue of low mammographic density. Conclusions Constitutive expression of CCL2 by the mouse mammary epithelium induces a state of low level chronic inflammation that increases stromal density and elevates cancer risk. We propose that CCL2-driven inflammation contributes to the increased risk of breast cancer observed in women with high mammographic density.
Collapse
Affiliation(s)
- Xuan Sun
- Discipline of Obstetrics and Gynaecology, School of Medicine, University of Adelaide, Adelaide, Australia.,The Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Danielle J Glynn
- Discipline of Obstetrics and Gynaecology, School of Medicine, University of Adelaide, Adelaide, Australia.,The Robinson Research Institute, University of Adelaide, Adelaide, Australia.,Discipline of Surgery, School of Medicine, The Queen Elizabeth Hospital, University of Adelaide, DX465702, 28 Woodville Rd, Woodville, 5011, Australia
| | - Leigh J Hodson
- The Robinson Research Institute, University of Adelaide, Adelaide, Australia.,Discipline of Surgery, School of Medicine, The Queen Elizabeth Hospital, University of Adelaide, DX465702, 28 Woodville Rd, Woodville, 5011, Australia
| | - Cecilia Huo
- The University of Melbourne Department of Surgery, St Vincent's Hospital Melbourne, Fitzroy, Australia
| | - Kara Britt
- Metastasis Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Erik W Thompson
- The University of Melbourne Department of Surgery, St Vincent's Hospital Melbourne, Fitzroy, Australia.,Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology and Translational Research Institute, Queensland, Australia
| | - Lucy Woolford
- School of Veterinary Sciences, University of Adelaide, Roseworthy, SA, Australia
| | - Andreas Evdokiou
- Discipline of Surgery, School of Medicine, The Queen Elizabeth Hospital, University of Adelaide, DX465702, 28 Woodville Rd, Woodville, 5011, Australia
| | - Jeffrey W Pollard
- MRC and University of Edinburgh Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Sarah A Robertson
- Discipline of Obstetrics and Gynaecology, School of Medicine, University of Adelaide, Adelaide, Australia.,The Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Wendy V Ingman
- The Robinson Research Institute, University of Adelaide, Adelaide, Australia. .,Discipline of Surgery, School of Medicine, The Queen Elizabeth Hospital, University of Adelaide, DX465702, 28 Woodville Rd, Woodville, 5011, Australia.
| |
Collapse
|
117
|
Sathi GA, Farahat M, Hara ES, Taketa H, Nagatsuka H, Kuboki T, Matsumoto T. MCSF orchestrates branching morphogenesis in developing submandibular gland tissue. J Cell Sci 2017; 130:1559-1569. [DOI: 10.1242/jcs.196907] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 02/21/2017] [Indexed: 12/16/2022] Open
Abstract
The importance of macrophages in tissue development and regeneration have been strongly emphasized. However, the specific roles of macrophage colony-stimulating factor (MCSF), the key regulator of macrophage differentiation, in glandular tissue development have been unexplored. Here, we disclose new macrophage-independent roles of MCSF in tissue development. We initially found that MCSF is markedly upregulated at embryonic day E13.5, at a stage preceding the colonization of macrophages (at E15.5) in mouse submandibular gland (SMG) tissue. Surprisingly, MCSF-induced branching morphogenesis was based on a direct effect on epithelial cells, as well as indirectly, by modulating the expression of major growth factors of SMG growth, FGF7 and FGF10, via the phosphoinositide 3-kinase (PI3K) pathway. Additionally, given the importance of neurons in SMG organogenesis, MCSF-induced SMG growth was associated with regulation of neurturin expression and neuronal network development during early SMG development in an in vitro organogenesis model as well as in vivo. These results indicate that MCSF plays pleiotropic roles and is an important regulator of early SMG morphogenesis.
Collapse
Affiliation(s)
- Gulsan Ara Sathi
- Department of Biomaterials, Okayama University, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
| | - Mahmoud Farahat
- Department of Biomaterials, Okayama University, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
| | - Emilio Satoshi Hara
- Department of Biomaterials, Okayama University, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
| | - Hiroaki Taketa
- Center for the Development of Medical and Health Care Education, Okayama University, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
| | - Hitoshi Nagatsuka
- Department of Oral Pathology and Medicine, Okayama University, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
| | - Takuo Kuboki
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
| | - Takuya Matsumoto
- Department of Biomaterials, Okayama University, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
| |
Collapse
|
118
|
McBryan J, Howlin J. Pubertal Mammary Gland Development: Elucidation of In Vivo Morphogenesis Using Murine Models. Methods Mol Biol 2017; 1501:77-114. [PMID: 27796948 DOI: 10.1007/978-1-4939-6475-8_3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
During the past 25 years, the combination of increasingly sophisticated gene targeting technology with transplantation techniques has allowed researchers to address a wide array of questions about postnatal mammary gland development. These in turn have significantly contributed to our knowledge of other branched epithelial structures. This review chapter highlights a selection of the mouse models exhibiting a pubertal mammary gland phenotype with a focus on how they have contributed to our overall understanding of in vivo mammary morphogenesis. We discuss mouse models that have enabled us to assign functions to particular genes and proteins and, more importantly, have determined when and where these factors are required for completion of ductal outgrowth and branch patterning. The reason for the success of the mouse mammary gland model is undoubtedly the suitability of the postnatal mammary gland to experimental manipulation. The gland itself is very amenable to investigation and the combination of genetic modification with accessibility to the tissue has allowed an impressive number of studies to inform biology. Excision of the rudimentary epithelial structure postnatally allows genetically modified tissue to be readily transplanted into wild type stroma or vice versa, and has thus defined the contribution of each compartment to particular phenotypes. Similarly, whole gland transplantation has been used to definitively discern local effects from indirect systemic effects of various growth factors and hormones. While appreciative of the power of these tools and techniques, we are also cognizant of some of their limitations, and we discuss some shortcomings and future strategies that can overcome them.
Collapse
Affiliation(s)
- Jean McBryan
- Department of Molecular Medicine Royal College of Surgeons in Ireland Education and Research Centre, Beaumont Hospital, Dublin, 9, Ireland
| | - Jillian Howlin
- Division of Oncology-Pathology, Lund University Cancer Center/Medicon Village, Building 404:B2, Scheelevägen 2, 223 81, Lund, Sweden.
| |
Collapse
|
119
|
Wilson GJ, Hewit KD, Pallas KJ, Cairney CJ, Lee KM, Hansell CA, Stein T, Graham GJ. Atypical chemokine receptor ACKR2 controls branching morphogenesis in the developing mammary gland. Development 2017; 144:74-82. [PMID: 27888192 PMCID: PMC5278629 DOI: 10.1242/dev.139733] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 11/07/2016] [Indexed: 02/01/2023]
Abstract
Macrophages are important regulators of branching morphogenesis during development and postnatally in the mammary gland. Regulation of macrophage dynamics during these processes can therefore have a profound impact on development. We demonstrate here that the developing mammary gland expresses high levels of inflammatory CC-chemokines, which are essential in vivo regulators of macrophage migration. We further demonstrate that the atypical chemokine receptor ACKR2, which scavenges inflammatory CC-chemokines, is differentially expressed during mammary gland development. We have previously shown that ACKR2 regulates macrophage dynamics during lymphatic vessel development. Here, we extend these observations to reveal a novel role for ACKR2 in regulating the postnatal development of the mammary gland. Specifically, we show that Ackr2-/- mice display precocious mammary gland development. This is associated with increased macrophage recruitment to the developing gland and increased density of the ductal epithelial network. These data demonstrate that ACKR2 is an important regulator of branching morphogenesis in diverse biological contexts and provide the first evidence of a role for chemokines and their receptors in postnatal development processes.
Collapse
Affiliation(s)
- Gillian J Wilson
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TT, UK
| | - Kay D Hewit
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TT, UK
| | - Kenneth J Pallas
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TT, UK
| | - Claire J Cairney
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Kit M Lee
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TT, UK
| | - Christopher A Hansell
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TT, UK
| | - Torsten Stein
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Gerard J Graham
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TT, UK
| |
Collapse
|
120
|
Mescher AL, Neff AW, King MW. Inflammation and immunity in organ regeneration. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 66:98-110. [PMID: 26891614 DOI: 10.1016/j.dci.2016.02.015] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/19/2016] [Accepted: 02/09/2016] [Indexed: 06/05/2023]
Abstract
The ability of vertebrates to regenerate amputated appendages is increasingly well-understood at the cellular level. Cells mediating an innate immune response and inflammation in the injured tissues are a prominent feature of the limb prior to formation of a regeneration blastema, with macrophage activity necessary for blastema growth and successful development of the new limb. Studies involving either anti-inflammatory or pro-inflammatory agents suggest that the local inflammation produced by injury and its timely resolution are both important for regeneration, with blastema patterning inhibited in the presence of unresolved inflammation. Various experiments with Xenopus larvae at stages where regenerative competence is declining show improved digit formation after treatment with certain immunosuppressive, anti-inflammatory, or antioxidant agents. Similar work with the larval Xenopus tail has implicated adaptive immunity with regenerative competence and suggests a requirement for regulatory T cells in regeneration, which also occurs in many systems of tissue regeneration. Recent analyses of the human nail organ indicate a capacity for local immune tolerance, suggesting roles for adaptive immunity in the capacity for mammalian appendage regeneration. New information and better understanding regarding the neuroendocrine-immune axis in the response to stressors, including amputation, suggest additional approaches useful for investigating effects of the immune system during repair and regeneration.
Collapse
Affiliation(s)
- Anthony L Mescher
- Center for Developmental and Regenerative Biology; Indiana University School of Medicine - Bloomington, USA.
| | - Anton W Neff
- Center for Developmental and Regenerative Biology; Indiana University School of Medicine - Bloomington, USA.
| | - Michael W King
- Center for Developmental and Regenerative Biology; Indiana University School of Medicine - Terre Haute, USA.
| |
Collapse
|
121
|
Abstract
Historically, eosinophils have been considered as end-stage cells involved in host protection against parasitic infection and in the mechanisms of hypersensitivity. However, later studies have shown that this multifunctional cell is also capable of producing immunoregulatory cytokines and soluble mediators and is involved in tissue homeostasis and modulation of innate and adaptive immune responses. In this review, we summarize the biology of eosinophils, including the function and molecular mechanisms of their granule proteins, cell surface markers, mediators, and pathways, and present comprehensive reviews of research updates on the genetics and epigenetics of eosinophils. We describe recent advances in the development of epigenetics of eosinophil-related diseases, especially in asthma. Likewise, recent studies have provided us with a more complete appreciation of how eosinophils contribute to the pathogenesis of various diseases, including hypereosinophilic syndrome (HES). Over the past decades, the definition and criteria of HES have been evolving with the progress of our understanding of the disease and some aspects of this disease still remain controversial. We also review recent updates on the genetic and molecular mechanisms of HES, which have spurred dramatic developments in the clinical strategies of diagnosis and treatment for this heterogeneous group of diseases. The conclusion from this review is that the biology of eosinophils provides significant insights as to their roles in health and disease and, furthermore, demonstrates that a better understanding of eosinophil will accelerate the development of new therapeutic strategies for patients.
Collapse
|
122
|
Peuhu E, Kaukonen R, Lerche M, Saari M, Guzmán C, Rantakari P, De Franceschi N, Wärri A, Georgiadou M, Jacquemet G, Mattila E, Virtakoivu R, Liu Y, Attieh Y, Silva KA, Betz T, Sundberg JP, Salmi M, Deugnier MA, Eliceiri KW, Ivaska J. SHARPIN regulates collagen architecture and ductal outgrowth in the developing mouse mammary gland. EMBO J 2016; 36:165-182. [PMID: 27974362 DOI: 10.15252/embj.201694387] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 10/28/2016] [Accepted: 10/28/2016] [Indexed: 11/09/2022] Open
Abstract
SHARPIN is a widely expressed multifunctional protein implicated in cancer, inflammation, linear ubiquitination and integrin activity inhibition; however, its contribution to epithelial homeostasis remains poorly understood. Here, we examined the role of SHARPIN in mammary gland development, a process strongly regulated by epithelial-stromal interactions. Mice lacking SHARPIN expression in all cells (Sharpincpdm), and mice with a stromal (S100a4-Cre) deletion of Sharpin, have reduced mammary ductal outgrowth during puberty. In contrast, Sharpincpdm mammary epithelial cells transplanted in vivo into wild-type stroma, fully repopulate the mammary gland fat pad, undergo unperturbed ductal outgrowth and terminal differentiation. Thus, SHARPIN is required in mammary gland stroma during development. Accordingly, stroma adjacent to invading mammary ducts of Sharpincpdm mice displayed reduced collagen arrangement and extracellular matrix (ECM) stiffness. Moreover, Sharpincpdm mammary gland stromal fibroblasts demonstrated defects in collagen fibre assembly, collagen contraction and degradation in vitro Together, these data imply that SHARPIN regulates the normal invasive mammary gland branching morphogenesis in an epithelial cell extrinsic manner by controlling the organisation of the stromal ECM.
Collapse
Affiliation(s)
- Emilia Peuhu
- Centre for Biotechnology, University of Turku, Turku, Finland
| | - Riina Kaukonen
- Centre for Biotechnology, University of Turku, Turku, Finland
| | - Martina Lerche
- Centre for Biotechnology, University of Turku, Turku, Finland
| | - Markku Saari
- Centre for Biotechnology, University of Turku, Turku, Finland
| | - Camilo Guzmán
- Centre for Biotechnology, University of Turku, Turku, Finland
| | - Pia Rantakari
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | | | - Anni Wärri
- Centre for Biotechnology, University of Turku, Turku, Finland
| | | | | | - Elina Mattila
- Centre for Biotechnology, University of Turku, Turku, Finland
| | | | - Yuming Liu
- Department of Biomedical Engineering, Laboratory for Optical and Computational Instrumentation (LOCI), University of Wisconsin at Madison, Madison, WI, USA
| | - Youmna Attieh
- Institut Curie, Paris Sciences et Lettres Research University, Paris, France
| | | | - Timo Betz
- Institut Curie, Paris Sciences et Lettres Research University, Paris, France.,Center for Molecular Biology of Inflammation, Cells-in-Motion Cluster of Excellence, Institute of Cell Biology, Münster University, Münster, Germany
| | | | - Marko Salmi
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - Marie-Ange Deugnier
- Institut Curie, Paris Sciences et Lettres Research University, Paris, France.,Institut Curie, CNRS, UMR144, Paris, France
| | - Kevin W Eliceiri
- Department of Biomedical Engineering, Laboratory for Optical and Computational Instrumentation (LOCI), University of Wisconsin at Madison, Madison, WI, USA
| | - Johanna Ivaska
- Centre for Biotechnology, University of Turku, Turku, Finland .,Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| |
Collapse
|
123
|
Dobrolecki LE, Airhart SD, Alferez DG, Aparicio S, Behbod F, Bentires-Alj M, Brisken C, Bult CJ, Cai S, Clarke RB, Dowst H, Ellis MJ, Gonzalez-Suarez E, Iggo RD, Kabos P, Li S, Lindeman GJ, Marangoni E, McCoy A, Meric-Bernstam F, Piwnica-Worms H, Poupon MF, Reis-Filho J, Sartorius CA, Scabia V, Sflomos G, Tu Y, Vaillant F, Visvader JE, Welm A, Wicha MS, Lewis MT. Patient-derived xenograft (PDX) models in basic and translational breast cancer research. Cancer Metastasis Rev 2016; 35:547-573. [PMID: 28025748 PMCID: PMC5396460 DOI: 10.1007/s10555-016-9653-x] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Patient-derived xenograft (PDX) models of a growing spectrum of cancers are rapidly supplanting long-established traditional cell lines as preferred models for conducting basic and translational preclinical research. In breast cancer, to complement the now curated collection of approximately 45 long-established human breast cancer cell lines, a newly formed consortium of academic laboratories, currently from Europe, Australia, and North America, herein summarizes data on over 500 stably transplantable PDX models representing all three clinical subtypes of breast cancer (ER+, HER2+, and "Triple-negative" (TNBC)). Many of these models are well-characterized with respect to genomic, transcriptomic, and proteomic features, metastatic behavior, and treatment response to a variety of standard-of-care and experimental therapeutics. These stably transplantable PDX lines are generally available for dissemination to laboratories conducting translational research, and contact information for each collection is provided. This review summarizes current experiences related to PDX generation across participating groups, efforts to develop data standards for annotation and dissemination of patient clinical information that does not compromise patient privacy, efforts to develop complementary data standards for annotation of PDX characteristics and biology, and progress toward "credentialing" of PDX models as surrogates to represent individual patients for use in preclinical and co-clinical translational research. In addition, this review highlights important unresolved questions, as well as current limitations, that have hampered more efficient generation of PDX lines and more rapid adoption of PDX use in translational breast cancer research.
Collapse
Affiliation(s)
- Lacey E. Dobrolecki
- The Lester and Sue Smith Breast Center, Departments of Molecular and Cellular Biology and Radiology, Baylor College of Medicine, Houston TX 77030,
| | | | - Denis G. Alferez
- Breast Cancer Now Research Unit, Division of Molecular and Clinical Cancer Studies, Manchester Cancer Research Centre, University of Manchester, Wilmslow Road, Manchester, M21 4QL, UK,
| | - Samuel Aparicio
- Dept. Path & Lab Medicine, BC Cancer Agency, 675 W10th Avenue, Vancouver V6R 3A6, Canada,
| | - Fariba Behbod
- Department of Pathology, University of Kansas Medical Center, 3901 Rainbow Blvd, WHE 1005B, Kansas City, KS 66160,
| | - Mohamed Bentires-Alj
- Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
- Lab 306, Hebelstrasse 20, CH-4031 Basel, Switzerland,
| | - Cathrin Brisken
- ISREC - Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), SV2.832 Station 19, CH-1015 Lausanne, Switzerland. Phone +41 (0)21 693 07 81, Sec: +41 (0)21 693 07 62, Fax +41 (0)21 693 07 40,
| | | | - Shirong Cai
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030,
| | - Robert B. Clarke
- Breast Cancer Now Research Unit, Division of Molecular and Clinical Cancer Studies, Manchester Cancer Research Centre, University of Manchester, Wilmslow Road, Manchester, M21 4QL, UK,
| | - Heidi Dowst
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston TX 77030,
| | - Matthew J. Ellis
- The Lester and Sue Smith Breast Center, Departments of Molecular and Cellular Biology and Radiology, Baylor College of Medicine, Houston TX 77030,
| | - Eva Gonzalez-Suarez
- Cancer Epigenetics and Biology Program, PEBC, Bellvitge Institute for Biomedical Research, IDIBELL, Av.Gran Via de L'Hospitalet, 199 – 203, 08908 L'Hospitalet de Llobregat, Barcelona, Spain, , Phone: +34 932607347, Fax: +34 932607139
| | - Richard D. Iggo
- INSERM U1218, Bergonié Cancer Institute, 229 cours de l'Argonne, 33076 Bordeaux, France,
| | - Peter Kabos
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045,
| | - Shunqiang Li
- Department of Internal Medicine, Washington University, St. Louis, MO 63130, Tel. 314-747-9311,
| | - Geoffrey J. Lindeman
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC 3010, Australia
- Familial Cancer Centre, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre. Grattan St, Parkville, VIC 3050, Australia,
| | - Elisabetta Marangoni
- Translational Research Department, Institut Curie, 26, rue d’Ulm, 75005 Paris - FRANCE,
| | - Aaron McCoy
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030,
| | - Funda Meric-Bernstam
- Departments of Investigational Cancer Therapeutics and Breast Surgical Oncology, UT M. D. Anderson Cancer Center, Houston TX 77030,
| | - Helen Piwnica-Worms
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030,
| | - Marie-France Poupon
- Founder and Scientific Advisor, Xentech SA, Genepole, 4 rue Pierre Fontaine, 91000 Evry, France,
| | - Jorge Reis-Filho
- Director of Experimental Pathology, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
- Affiliate Member, Human Oncology and Pathogenesis Program, and Center for Computational Biology, Memorial Sloan Kettering Cancer Center, New York, NY,
| | - Carol A. Sartorius
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045,
| | - Valentina Scabia
- ISREC - Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), SV2.832 Station 19, CH-1015 Lausanne, Switzerland,
| | - George Sflomos
- ISREC - Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), SV2.832 Station 19, CH-1015 Lausanne, Switzerland.
| | - Yizheng Tu
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030,
| | - François Vaillant
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia,
| | - Jane E. Visvader
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia,
| | - Alana Welm
- Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT 84112,
| | - Max S. Wicha
- Madeline and Sidney Forbes Professor of Oncology, Director, Forbes Institute for Cancer Discovery, NCRC 26-335S, SPC 2800, 2800 Plymouth Rd., Ann Arbor, MI 48109-2800, Phone: (734)763-1744, Fax: (734)764-1228, http://www.med.umich.edu/wicha-lab/index.html,
| | - Michael T. Lewis
- The Lester and Sue Smith Breast Center, Departments of Molecular and Cellular Biology and Radiology, Baylor College of Medicine, Houston TX 77030, , TEL: 713-798-3296, FAX: 713-798-1659
| |
Collapse
|
124
|
Abstract
Macrophages are found in all tissues and regulate tissue morphogenesis during development through trophic and scavenger functions. The colony stimulating factor-1 (CSF-1) receptor (CSF-1R) is the major regulator of tissue macrophage development and maintenance. In combination with receptor activator of nuclear factor κB (RANK), the CSF-1R also regulates the differentiation of the bone-resorbing osteoclast and controls bone remodeling during embryonic and early postnatal development. CSF-1R-regulated macrophages play trophic and remodeling roles in development. Outside the mononuclear phagocytic system, the CSF-1R directly regulates neuronal survival and differentiation, the development of intestinal Paneth cells and of preimplantation embryos, as well as trophoblast innate immune function. Consistent with the pleiotropic roles of the receptor during development, CSF-1R deficiency in most mouse strains causes embryonic or perinatal death and the surviving mice exhibit multiple developmental and functional deficits. The CSF-1R is activated by two dimeric glycoprotein ligands, CSF-1, and interleukin-34 (IL-34). Homozygous Csf1-null mutations phenocopy most of the deficits of Csf1r-null mice. In contrast, Il34-null mice have no gross phenotype, except for decreased numbers of Langerhans cells and microglia, indicating that CSF-1 plays the major developmental role. Homozygous inactivating mutations of the Csf1r or its ligands have not been reported in man. However, heterozygous inactivating mutations in the Csf1r lead to a dominantly inherited adult-onset progressive dementia, highlighting the importance of CSF-1R signaling in the brain.
Collapse
Affiliation(s)
- Violeta Chitu
- Albert Einstein College of Medicine, Bronx, NY, United States
| | | |
Collapse
|
125
|
Sapi E. The Role of CSF-1 in Normal Physiology of Mammary Gland and Breast Cancer: An Update. Exp Biol Med (Maywood) 2016; 229:1-11. [PMID: 14709771 DOI: 10.1177/153537020422900101] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Colony stimulating factor (CSF-1) and its receptor (CSF-1R, product of c-fms proto-oncogene) were initially implicated as essential for normal monocyte development as well as for trophoblastic implantation. However, studies have demonstrated that CSF-1 and CSF-1R have additional roles in mammary gland development during pregnancy and lactation. This apparent role for CSF-1/CSF-1R in normal mammary gland development is very intriguing because this receptor/ligand pair has also been found to be important in the biology of breast cancer in which abnormal expression of CSF-1 and its receptor correlates with tumor cell invasiveness and adverse clinical prognosis. Recent findings also implicate tumor-produced CSF-1 in promotion of bone metastasis in breast cancer, and a certain membrane-associated form of CSF-1 appears to induce immunity against tumors. This review aims to summarize recent findings on the role of CSF-1 and its receptor in normal and neoplastic mammary development that may elucidate potential relationships of growth factor–induced biological changes in the breast during pregnancy and tumor progression.
Collapse
Affiliation(s)
- Eva Sapi
- Department of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut 06520-8040, USA.
| |
Collapse
|
126
|
Gangwar RS, Landolina N, Arpinati L, Levi-Schaffer F. Mast cell and eosinophil surface receptors as targets for anti-allergic therapy. Pharmacol Ther 2016; 170:37-63. [PMID: 27773785 DOI: 10.1016/j.pharmthera.2016.10.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Roopesh Singh Gangwar
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Nadine Landolina
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Ludovica Arpinati
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Francesca Levi-Schaffer
- Pharmacology & Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel.
| |
Collapse
|
127
|
Schlitzer A, Schultze JL. Tissue‐resident macrophages — how to humanize our knowledge. Immunol Cell Biol 2016; 95:173-177. [DOI: 10.1038/icb.2016.82] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 08/29/2016] [Accepted: 08/29/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Andreas Schlitzer
- Department of Genomics and Immunoregulation, Life and Medical Science Institute, University of Bonn Bonn Germany
- Single Cell Genomics and Epigenomics Unit, German Center for Neurodegenerative Diseases, University of Bonn Bonn Germany
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR) Singapore Singapore
| | - Joachim L Schultze
- Department of Genomics and Immunoregulation, Life and Medical Science Institute, University of Bonn Bonn Germany
- Single Cell Genomics and Epigenomics Unit, German Center for Neurodegenerative Diseases, University of Bonn Bonn Germany
| |
Collapse
|
128
|
Li L, Chen J, Xiong G, St Clair DK, Xu W, Xu R. Increased ROS production in non-polarized mammary epithelial cells induces monocyte infiltration in 3D culture. J Cell Sci 2016; 130:190-202. [PMID: 27656113 DOI: 10.1242/jcs.186031] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 08/17/2016] [Indexed: 02/01/2023] Open
Abstract
Loss of epithelial cell polarity promotes cell invasion and cancer dissemination. Therefore, identification of factors that disrupt polarized acinar formation is crucial. Reactive oxygen species (ROS) drive cancer progression and promote inflammation. Here, we show that the non-polarized breast cancer cell line T4-2 generates significantly higher ROS levels than polarized S1 and T4R cells in three-dimensional (3D) culture, accompanied by induction of the nuclear factor κB (NF-κB) pathway and cytokine expression. Minimizing ROS in T4-2 cells with antioxidants reestablished basal polarity and inhibited cell proliferation. Introducing constitutively activated RAC1 disrupted cell polarity and increased ROS levels, indicating that RAC1 is a crucial regulator that links cell polarity and ROS generation. We also linked monocyte infiltration with disruption of polarized acinar structure using a 3D co-culture system. Gain- and loss-of-function experiments demonstrated that increased ROS in non-polarized cells is necessary and sufficient to enhance monocyte recruitment. ROS also induced cytokine expression and NF-κB activity. These results suggest that increased ROS production in mammary epithelial cell leads to disruption of cell polarity and promotes monocyte infiltration.
Collapse
Affiliation(s)
- Linzhang Li
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, Jilin Province 130021, China.,Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Jie Chen
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Gaofeng Xiong
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Daret K St Clair
- Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Wei Xu
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Ren Xu
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA .,Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
129
|
Mesnil C, Raulier S, Paulissen G, Xiao X, Birrell MA, Pirottin D, Janss T, Starkl P, Ramery E, Henket M, Schleich FN, Radermecker M, Thielemans K, Gillet L, Thiry M, Belvisi MG, Louis R, Desmet C, Marichal T, Bureau F. Lung-resident eosinophils represent a distinct regulatory eosinophil subset. J Clin Invest 2016; 126:3279-95. [PMID: 27548519 DOI: 10.1172/jci85664] [Citation(s) in RCA: 358] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 06/09/2016] [Indexed: 12/21/2022] Open
Abstract
Increases in eosinophil numbers are associated with infection and allergic diseases, including asthma, but there is also evidence that eosinophils contribute to homeostatic immune processes. In mice, the normal lung contains resident eosinophils (rEos), but their function has not been characterized. Here, we have reported that steady-state pulmonary rEos are IL-5-independent parenchymal Siglec-FintCD62L+CD101lo cells with a ring-shaped nucleus. During house dust mite-induced airway allergy, rEos features remained unchanged, and rEos were accompanied by recruited inflammatory eosinophils (iEos), which were defined as IL-5-dependent peribronchial Siglec-FhiCD62L-CD101hi cells with a segmented nucleus. Gene expression analyses revealed a more regulatory profile for rEos than for iEos, and correspondingly, mice lacking lung rEos showed an increase in Th2 cell responses to inhaled allergens. Such elevation of Th2 responses was linked to the ability of rEos, but not iEos, to inhibit the maturation, and therefore the pro-Th2 function, of allergen-loaded DCs. Finally, we determined that the parenchymal rEos found in nonasthmatic human lungs (Siglec-8+CD62L+IL-3Rlo cells) were phenotypically distinct from the iEos isolated from the sputa of eosinophilic asthmatic patients (Siglec-8+CD62LloIL-3Rhi cells), suggesting that our findings in mice are relevant to humans. In conclusion, our data define lung rEos as a distinct eosinophil subset with key homeostatic functions.
Collapse
|
130
|
Shinozawa T, Yoshikawa HY, Takebe T. Reverse engineering liver buds through self-driven condensation and organization towards medical application. Dev Biol 2016; 420:221-229. [PMID: 27364470 DOI: 10.1016/j.ydbio.2016.06.036] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 05/24/2016] [Accepted: 06/25/2016] [Indexed: 12/15/2022]
Abstract
The self-organizing tissue-based approach coupled with induced pluripotent stem (iPS) cell technology is evolving as a promising field for designing organoids in culture and is expected to achieve valuable practical outcomes in regenerative medicine and drug development. Organoids show properties of functional organs and represent an alternative to cell models in conventional two-dimensional differentiation platforms; moreover, organoids can be used to investigate mechanisms of development and disease, drug discovery and toxicity assessment. Towards a more complex and advanced organoid model, it is essential to incorporate multiple cell lineages including developing vessels. Using a self-condensation method, we recently demonstrated self-organizing "organ buds" of diverse systems together with human mesenchymal and endothelial progenitors, proposing a new reverse engineering method to generate a more complex organoid structure. In this section, we review characters of organ bud technology based on two important principles: self-condensation and self-organization focusing on liver bud as an example, and discuss their practicality in regenerative medicine and potential as research tools for developmental biology and drug discovery.
Collapse
Affiliation(s)
- Tadahiro Shinozawa
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Hiroshi Y Yoshikawa
- Department of Chemistry, Saitama University, Shimo-okubo 255, Sakura-ku, Saitama 338-8570, Japan.
| | - Takanori Takebe
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan; PRESTO, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan.
| |
Collapse
|
131
|
Yu YRA, O’Koren EG, Hotten DF, Kan MJ, Kopin D, Nelson ER, Que L, Gunn MD. A Protocol for the Comprehensive Flow Cytometric Analysis of Immune Cells in Normal and Inflamed Murine Non-Lymphoid Tissues. PLoS One 2016; 11:e0150606. [PMID: 26938654 PMCID: PMC4777539 DOI: 10.1371/journal.pone.0150606] [Citation(s) in RCA: 264] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 02/16/2016] [Indexed: 11/18/2022] Open
Abstract
Flow cytometry is used extensively to examine immune cells in non-lymphoid tissues. However, a method of flow cytometric analysis that is both comprehensive and widely applicable has not been described. We developed a protocol for the flow cytometric analysis of non-lymphoid tissues, including methods of tissue preparation, a 10-fluorochrome panel for cell staining, and a standardized gating strategy, that allows the simultaneous identification and quantification of all major immune cell types in a variety of normal and inflamed non-lymphoid tissues. We demonstrate that our basic protocol minimizes cell loss, reliably distinguishes macrophages from dendritic cells (DC), and identifies all major granulocytic and mononuclear phagocytic cell types. This protocol is able to accurately quantify 11 distinct immune cell types, including T cells, B cells, NK cells, neutrophils, eosinophils, inflammatory monocytes, resident monocytes, alveolar macrophages, resident/interstitial macrophages, CD11b- DC, and CD11b+ DC, in normal lung, heart, liver, kidney, intestine, skin, eyes, and mammary gland. We also characterized the expression patterns of several commonly used myeloid and macrophage markers. This basic protocol can be expanded to identify additional cell types such as mast cells, basophils, and plasmacytoid DC, or perform detailed phenotyping of specific cell types. In examining models of primary and metastatic mammary tumors, this protocol allowed the identification of several distinct tumor associated macrophage phenotypes, the appearance of which was highly specific to individual tumor cell lines. This protocol provides a valuable tool to examine immune cell repertoires and follow immune responses in a wide variety of tissues and experimental conditions.
Collapse
Affiliation(s)
- Yen-Rei A. Yu
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| | - Emily G. O’Koren
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Danielle F. Hotten
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Matthew J. Kan
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - David Kopin
- School of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Erik R. Nelson
- Department of Molecular & Integrative Physiology, University of Illinois, Champaign, Illinois, United States of America
| | - Loretta Que
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Michael D. Gunn
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
132
|
Macrophages: Regulators of the Inflammatory Microenvironment during Mammary Gland Development and Breast Cancer. Mediators Inflamm 2016; 2016:4549676. [PMID: 26884646 PMCID: PMC4739263 DOI: 10.1155/2016/4549676] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 12/21/2015] [Indexed: 12/22/2022] Open
Abstract
Macrophages are critical mediators of inflammation and important regulators of developmental processes. As a key phagocytic cell type, macrophages evolved as part of the innate immune system to engulf and process cell debris and pathogens. Macrophages produce factors that act directly on their microenvironment and also bridge innate immune responses to the adaptive immune system. Resident macrophages are important for acting as sensors for tissue damage and maintaining tissue homeostasis. It is now well-established that macrophages are an integral component of the breast tumor microenvironment, where they contribute to tumor growth and progression, likely through many of the mechanisms that are utilized during normal wound healing responses. Because macrophages contribute to normal mammary gland development and breast cancer growth and progression, this review will discuss both resident mammary gland macrophages and tumor-associated macrophages with an emphasis on describing how macrophages interact with their surrounding environment during normal development and in the context of cancer.
Collapse
|
133
|
Abstract
The clinical relevance of the host immune system in breast cancer has long been unexplored. Studies developed over the past decade have highlighted the biological heterogeneity of breast cancer, prompting researchers to investigate whether the role of the immune system in this malignancy is similar across different molecular subtypes of the disease. The presence of high levels of lymphocytic infiltration has been consistently associated with a more-favourable prognosis in patients with early stage triple-negative and HER2-positive breast cancer. These infiltrates seem to reflect favourable host antitumour immune responses, suggesting that immune activation is important for improving survival outcomes. In this Review, we discuss the composition of the immune infiltrates observed in breast cancers, as well as data supporting the clinical relevance of host antitumour immunity, as represented by lymphocytic infiltration, and how this biomarker could be used in the clinical setting. We also discuss the rationale for enhancing immunity in breast cancer, including early data on the efficacy of T-cell checkpoint inhibition in this setting.
Collapse
|
134
|
Ribatti D, Crivellato E. The role of mast cell in tissue morphogenesis. Thymus, duodenum, and mammary gland as examples. Exp Cell Res 2015; 341:105-109. [PMID: 26615957 DOI: 10.1016/j.yexcr.2015.11.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/18/2015] [Accepted: 11/21/2015] [Indexed: 11/29/2022]
Abstract
Mast cells (MCs) are strategically located at host/environment interfaces like skin, airways, and gastro-intestinal and uro-genital tracts. MCs also populate connective tissues in association with blood and lymphatic vessels and nerves. MCs are absent in avascular tissues, such as mineralized bone, cartilage, and cornea. MCs have various functions and different functional subsets of MCs are encountered in different tissues. However, we do not' know exactly what is the physiological function of MC. Most of these functions are not essential for life, as various MC-deficient strains of mice and rats seems to have normal life spans. In this review article, we have reported and discussed the literature data concerning the role of MCs in tissue morphogenesis, and in particular their role in the development of thymus, duodenum, and mammary gland.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy; National Cancer Institute "Giovanni Paolo II", Bari, Italy.
| | - Enrico Crivellato
- Department of Experimental and Clinical Medicine, Section of Human Anatomy, University of Udine, Udine, Italy
| |
Collapse
|
135
|
Beaudry KL, Parsons CLM, Ellis SE, Akers RM. Localization and quantitation of macrophages, mast cells, and eosinophils in the developing bovine mammary gland. J Dairy Sci 2015; 99:796-804. [PMID: 26547646 DOI: 10.3168/jds.2015-9972] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 09/15/2015] [Indexed: 11/19/2022]
Abstract
Prepubertal mammary development involves elongation and branching of ducts and stromal tissue remodeling. This process is highly regulated and in mice is known to be affected by the presence of innate immune cells. Whether or not such immune cells are present or involved in bovine mammary development is unknown. For the first time, we determined the presence, location (relative to mammary ductal structures), and changes in numbers of eosinophils, mast cells, and macrophages in prepubertal bovine mammary tissue, and evaluated the effects of age, ovariectomy, and exogenous estrogen on numbers of each cell type. Chemical stains and immunofluorescence were used to identify the 3 cell types in formalin-fixed, paraffin-embedded mammary tissue from prepubertal female calves from 3 archived tissue sets. The ontogeny tissue set included samples of mammary tissue from female calves (n=4/wk) from birth to 6 wk of age. The ovary tissue set contained samples from ovary intact and ovariectomized heifers allowing us to investigate the influence of the ovaries on immune cells in the developing mammary gland in prepubertal heifers. Nineteen animals were intact or ovariectomized 30 d before sampling; they were 90, 120, or 150 d old at the time of sampling. A third tissue set, the estrogen set, allowed us to determine the effect of exogenous estrogen on innate immune cells in the gland. Eosinophils were identified via Luna staining, mast cells by May-Grunwald Giemsa staining, and macrophages with immunofluorescence. Key findings were that more eosinophils and mast cells were observed in near versus far stroma in the ontogeny and ovary tissue sets but not estrogen. More macrophages were observed in near versus far stroma in ontogeny animals. Eosinophils were more abundant in the younger animals, and fewer macrophages tended to be observed in ovariectomized heifers as compared with intact heifers and estrogen treatment resulted in a reduction in cell numbers. In summary, we show for the first time that innate immune cells are present in prepubertal bovine mammary tissue, localization varies by immune cell type, and abundance is related to proximity of epithelial structures and physiological state. We suggest a likely role for these cells in control of bovine mammary growth and ductal development.
Collapse
Affiliation(s)
- K L Beaudry
- Department of Dairy Sciences, Virginia Polytechnic Institute and State University, Blacksburg 24061
| | - C L M Parsons
- Department of Dairy Sciences, Virginia Polytechnic Institute and State University, Blacksburg 24061
| | - S E Ellis
- Department of Dairy Sciences, Virginia Polytechnic Institute and State University, Blacksburg 24061
| | - R M Akers
- Department of Dairy Sciences, Virginia Polytechnic Institute and State University, Blacksburg 24061.
| |
Collapse
|
136
|
Berryhill GE, Trott JF, Hovey RC. Mammary gland development--It's not just about estrogen. J Dairy Sci 2015; 99:875-83. [PMID: 26506542 DOI: 10.3168/jds.2015-10105] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 09/02/2015] [Indexed: 12/22/2022]
Abstract
The mammary gland (MG) is one of a few organs that undergoes most of its growth after birth. Much of this development occurs concurrently with specific reproductive states, such that the ultimate goal of milk synthesis and secretion is coordinated with the nutritional requirements of the neonate. Central to the reproductive-MG axis is its endocrine regulation, and pivotal to this regulation is the ovarian secretion of estrogen (E). Indeed, it is widely accepted that estrogens are essential for growth of the MG to occur, both for ductal elongation during puberty and for alveolar development during gestation. As the factors regulating MG development continually come to light from the fields of developmental biology, lactation physiology, and breast cancer research, a growing body of evidence serves as a reminder that the MG are not as exclusively dependent on estrogens as might have been thought. The objective of this review is to summarize the state of information regarding our understanding of how estrogen (E) has been implicated as the key regulator of MG development, and to highlight some of the alternative E-independent mechanisms that have been discovered. In particular, we review our findings that dietary trans-10,cis-12 conjugated linoleic acid promotes ductal elongation and that the combination of progesterone (P) and prolactin (PRL) can stimulate branching morphogenesis in the absence of E. Ultimately, these examples stand as a healthy challenge to the question of just how important estrogens are for MG development. Answers to this question, in turn, increase our understanding of MG development across all mammals and the ways in which it can affect milk production.
Collapse
Affiliation(s)
- Grace E Berryhill
- Department of Animal Science, University of California-Davis, 2145 Meyer Hall, One Shields Avenue, Davis 95618
| | - Josephine F Trott
- Department of Animal Science, University of California-Davis, 2145 Meyer Hall, One Shields Avenue, Davis 95618
| | - Russell C Hovey
- Department of Animal Science, University of California-Davis, 2145 Meyer Hall, One Shields Avenue, Davis 95618.
| |
Collapse
|
137
|
Greenow KR, Smalley MJ. Overview of Genetically Engineered Mouse Models of Breast Cancer Used in Translational Biology and Drug Development. CURRENT PROTOCOLS IN PHARMACOLOGY 2015; 70:14.36.1-14.36.14. [PMID: 26331886 DOI: 10.1002/0471141755.ph1436s70] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Breast cancer is a heterogeneous condition with no single standard of treatment and no definitive method for determining whether a tumor will respond to therapy. The development of murine models that faithfully mimic specific human breast cancer subtypes is critical for the development of patient-specific treatments. While the artificial nature of traditional in vivo xenograft models used to characterize novel anticancer treatments has limited clinical predictive value, the development of genetically engineered mouse models (GEMMs) makes it possible to study the therapeutic responses in an intact microenvironment. GEMMs have proven to be an experimentally tractable platform for evaluating the efficacy of novel therapeutic combinations and for defining the mechanisms of acquired resistance. Described in this overview are several of the more popular breast cancer GEMMs, including details on their value in elucidating the molecular mechanisms of this disorder.
Collapse
Affiliation(s)
- Kirsty R Greenow
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff, United Kingdom
- Current Address: Propath UK Ltd., Hereford, United Kingdom
| | - Matthew J Smalley
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff, United Kingdom
- Corresponding Author:
| |
Collapse
|
138
|
Plaks V, Boldajipour B, Linnemann JR, Nguyen NH, Kersten K, Wolf Y, Casbon AJ, Kong N, van den Bijgaart RJE, Sheppard D, Melton AC, Krummel MF, Werb Z. Adaptive Immune Regulation of Mammary Postnatal Organogenesis. Dev Cell 2015; 34:493-504. [PMID: 26321127 DOI: 10.1016/j.devcel.2015.07.015] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 05/13/2015] [Accepted: 07/28/2015] [Indexed: 01/15/2023]
Abstract
Postnatal organogenesis occurs in an immune competent environment and is tightly controlled by interplay between positive and negative regulators. Innate immune cells have beneficial roles in postnatal tissue remodeling, but roles for the adaptive immune system are currently unexplored. Here we show that adaptive immune responses participate in the normal postnatal development of a non-lymphoid epithelial tissue. Since the mammary gland (MG) is the only organ developing predominantly after birth, we utilized it as a powerful system to study adaptive immune regulation of organogenesis. We found that antigen-mediated interactions between mammary antigen-presenting cells and interferon-γ (IFNγ)-producing CD4+ T helper 1 cells participate in MG postnatal organogenesis as negative regulators, locally orchestrating epithelial rearrangement. IFNγ then affects luminal lineage differentiation. This function of adaptive immune responses, regulating normal development, changes the paradigm for studying players of postnatal organogenesis and provides insights into immune surveillance and cancer transformation.
Collapse
Affiliation(s)
- Vicki Plaks
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Bijan Boldajipour
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jelena R Linnemann
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nguyen H Nguyen
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kelly Kersten
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yochai Wolf
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Amy-Jo Casbon
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Niwen Kong
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Dean Sheppard
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Andrew C Melton
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Zena Werb
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
139
|
Abstract
Macrophages form a heterogeneous group of hematopoietic cells that reside in tissues, where they are required to maintain organ integrity. Tissue macrophages contribute to tissue formation, metabolism, homeostasis, and repair. They have a unique ability to sense and respond to tissue damage. They serve as the first line of defense during infection and help promote immune tolerance in the steady state. Although most tissue macrophages share a high phagocytic and degradative potential, they are heterogeneous in origin, as well as in homeostatic function and response to insults. Here, we will discuss recent developments in our understanding of the origin of tissue macrophages and their functional specialization in tissues.
Collapse
Affiliation(s)
- Yonit Lavin
- Authors' Affiliation: Department of Oncological Science, The Tisch Cancer Institute and The Immunology Institute, Mount Sinai School of Medicine, New York, New York
| | | |
Collapse
|
140
|
Ratheesh A, Belyaeva V, Siekhaus DE. Drosophila immune cell migration and adhesion during embryonic development and larval immune responses. Curr Opin Cell Biol 2015. [PMID: 26210104 DOI: 10.1016/j.ceb.2015.07.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The majority of immune cells in Drosophila melanogaster are plasmatocytes; they carry out similar functions to vertebrate macrophages, influencing development as well as protecting against infection and cancer. Plasmatocytes, sometimes referred to with the broader term of hemocytes, migrate widely during embryonic development and cycle in the larvae between sessile and circulating positions. Here we discuss the similarities of plasmatocyte developmental migration and its functions to that of vertebrate macrophages, considering the recent controversy regarding the functions of Drosophila PDGF/VEGF related ligands. We also examine recent findings on the significance of adhesion for plasmatocyte migration in the embryo, as well as proliferation, trans-differentiation, and tumor responses in the larva. We spotlight parallels throughout to vertebrate immune responses.
Collapse
Affiliation(s)
| | - Vera Belyaeva
- IST Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | | |
Collapse
|
141
|
Lahmar Q, Keirsse J, Laoui D, Movahedi K, Van Overmeire E, Van Ginderachter JA. Tissue-resident versus monocyte-derived macrophages in the tumor microenvironment. Biochim Biophys Acta Rev Cancer 2015; 1865:23-34. [PMID: 26145884 DOI: 10.1016/j.bbcan.2015.06.009] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 06/25/2015] [Accepted: 06/26/2015] [Indexed: 12/12/2022]
Abstract
The tumor-promoting role of macrophages has been firmly established in most cancer types. However, macrophage identity has been a matter of debate, since several levels of complexity result in considerable macrophage heterogeneity. Ontogenically, tissue-resident macrophages derive from yolk sac progenitors which either directly or via a fetal liver monocyte intermediate differentiate into distinct macrophage types during embryogenesis and are maintained throughout life, while a disruption of the steady state mobilizes monocytes and instructs the formation of monocyte-derived macrophages. Histologically, the macrophage phenotype is heavily influenced by the tissue microenvironment resulting in molecularly and functionally distinct macrophages in distinct organs. Finally, a change in the tissue microenvironment as a result of infectious or sterile inflammation instructs different modes of macrophage activation. These considerations are relevant in the context of tumors, which can be considered as sites of chronic sterile inflammation encompassing subregions with distinct environmental conditions (for example, hypoxic versus normoxic). Here, we discuss existing evidence on the role of macrophage subpopulations in steady state tissue and primary tumors of the breast, lung, pancreas, brain and liver.
Collapse
Affiliation(s)
- Qods Lahmar
- Myeloid Cell Immunology Lab, VIB, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jiri Keirsse
- Myeloid Cell Immunology Lab, VIB, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Damya Laoui
- Myeloid Cell Immunology Lab, VIB, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kiavash Movahedi
- Myeloid Cell Immunology Lab, VIB, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eva Van Overmeire
- Myeloid Cell Immunology Lab, VIB, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jo A Van Ginderachter
- Myeloid Cell Immunology Lab, VIB, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
142
|
Morris DL. Minireview: Emerging Concepts in Islet Macrophage Biology in Type 2 Diabetes. Mol Endocrinol 2015; 29:946-62. [PMID: 26001058 DOI: 10.1210/me.2014-1393] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Chronic systemic inflammation is a hallmark feature of obesity and type 2 diabetes. Both resident and recruited islet macrophages contribute to the proinflammatory milieu of the diabetic islet. However, macrophages also appear to be critical for β-cell formation during development and support β-cell replication in experimental models of pancreas regeneration. In light of these findings, perhaps macrophages in the islet need to be viewed more as a fulcrum where deleterious inflammatory activation is balanced with beneficial tissue repair processes. Undoubtedly, defining the factors that contribute to the ontogeny, heterogeneity, and functionality of macrophages in normal, diseased, and regenerating islets will be necessary to determine whether that fulcrum can be moved to preserve functional β-cell mass in persons with diabetes. The intent of this review is to introduce the reader to emerging concepts of islet macrophage biology that may challenge the perception that macrophage accumulation in islets is merely a pathological feature of type 2 diabetes.
Collapse
Affiliation(s)
- David L Morris
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| |
Collapse
|
143
|
Abstract
The immune system, best known as the first line of defense against invading pathogens, is integral to tissue development, homeostasis, and wound repair. In recent years, there has been a growing appreciation that cellular and humoral components of the immune system also contribute to regeneration of damaged tissues, including limbs, skeletal muscle, heart, and the nervous system. Here, we discuss key findings that implicate inflammatory cells and their secreted factors in tissue replacement after injury via stem cells and other reparative mechanisms. We highlight clinical conditions that are amenable to immune-mediated regeneration and suggest immune targeting strategies for tissue regeneration.
Collapse
Affiliation(s)
- Arin B Aurora
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA
| | - Eric N Olson
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA.
| |
Collapse
|
144
|
Differential HIF-1α and HIF-2α Expression in Mammary Epithelial Cells during Fat Pad Invasion, Lactation, and Involution. PLoS One 2015; 10:e0125771. [PMID: 25955753 PMCID: PMC4425677 DOI: 10.1371/journal.pone.0125771] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 03/25/2015] [Indexed: 01/12/2023] Open
Abstract
The development and functional cycle of the mammary gland involves a number of processes that are caricatured by breast cancer cells during invasion and metastasis. Expression of the hypoxia-inducible transcription factors HIF-1 and HIF-2 has been associated with metastatic, poor prognosis, and high-grade breast cancers. Since hypoxia affects normal epithelial differentiation, we hypothesise that HIFs are important for normal breast epithelial development and regeneration as well as cancer initiation and progression. Here, we investigated the expression of the oxygen-sensitive HIF-alpha subunits during mouse mammary gland development, lactation, and involution. In breast epithelial cells, HIF-1α was expressed during early development, prior to cell polarisation. In contrast, expression of HIF-2α occurred later and was restricted to a subpopulation of luminal epithelial cells in the lactating gland. Mammary gland involution is a developmental stage that involves extensive tissue remodelling with cell death but survival of tissue stem/progenitor cells. At this stage, HIF-2α, but little HIF-1α, was expressed in CK14-positive epithelial cells. The temporal but differential expression of the HIF-alpha subunits during the mammary gland life cycle indicates that their expression is controlled by additional factors to hypoxia. Further functional studies of the roles of these proteins in the mammary gland and breast cancer are warranted.
Collapse
|
145
|
Abstract
Microglia are macrophages that colonize the brain during development to establish a resident population of professional phagocytes that protect against invading pathogens and contribute to brain development and homeostasis. As such, these cells sit at the interface between immunology and neurobiology. In addition to their key roles in brain physiology, microglia offer a great opportunity to address central questions in biology relating to how migrating cells find their positions in the embryo, adopt a behavior that is appropriate for that position, and interact with their local environment. We aim, in this review, to survey key recent advances in microglial research.
Collapse
Affiliation(s)
- Alessandra Maria Casano
- European Molecular Biology Laboratory, Developmental Biology Unit, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Francesca Peri
- European Molecular Biology Laboratory, Developmental Biology Unit, Meyerhofstrasse 1, 69117 Heidelberg, Germany.
| |
Collapse
|
146
|
Lee S, Hennigar SR, Alam S, Nishida K, Kelleher SL. Essential Role for Zinc Transporter 2 (ZnT2)-mediated Zinc Transport in Mammary Gland Development and Function during Lactation. J Biol Chem 2015; 290:13064-78. [PMID: 25851903 DOI: 10.1074/jbc.m115.637439] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Indexed: 01/28/2023] Open
Abstract
The zinc transporter ZnT2 (SLC30A2) imports zinc into vesicles in secreting mammary epithelial cells (MECs) and is critical for zinc efflux into milk during lactation. Recent studies show that ZnT2 also imports zinc into mitochondria and is expressed in the non-lactating mammary gland and non-secreting MECs, highlighting the importance of ZnT2 in general mammary gland biology. In this study we used nulliparous and lactating ZnT2-null mice and characterized the consequences on mammary gland development, function during lactation, and milk composition. We found that ZnT2 was primarily expressed in MECs and to a limited extent in macrophages in the nulliparous mammary gland and loss of ZnT2 impaired mammary expansion during development. Secondly, we found that lactating ZnT2-null mice had substantial defects in mammary gland architecture and MEC function during secretion, including fewer, condensed and disorganized alveoli, impaired Stat5 activation, and unpolarized MECs. Loss of ZnT2 led to reduced milk volume and milk containing less protein, fat, and lactose compared with wild-type littermates, implicating ZnT2 in the regulation of mammary differentiation and optimal milk production during lactation. Together, these results demonstrate that ZnT2-mediated zinc transport is critical for mammary gland function, suggesting that defects in ZnT2 not only reduce milk zinc concentration but may compromise breast health and increase the risk for lactation insufficiency in lactating women.
Collapse
Affiliation(s)
- Sooyeon Lee
- From the Interdisciplinary Graduate Physiology Program and Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, Departments of Cellular and Molecular Physiology
| | - Stephen R Hennigar
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Samina Alam
- Departments of Cellular and Molecular Physiology, Surgery, Penn State Hershey College of Medicine, Hershey, Pennsylvania 17033
| | - Keigo Nishida
- Laboratory for Homeostatic Network, RCAI, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), Yokohama 230-0045, Japan, and Laboratory of Immune Regulation, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka 513-8670, Japan
| | - Shannon L Kelleher
- From the Interdisciplinary Graduate Physiology Program and Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, Departments of Cellular and Molecular Physiology, Surgery, Penn State Hershey College of Medicine, Hershey, Pennsylvania 17033, Pharmacology, and
| |
Collapse
|
147
|
Inman JL, Robertson C, Mott JD, Bissell MJ. Mammary gland development: cell fate specification, stem cells and the microenvironment. Development 2015; 142:1028-42. [DOI: 10.1242/dev.087643] [Citation(s) in RCA: 279] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The development of the mammary gland is unique: the final stages of development occur postnatally at puberty under the influence of hormonal cues. Furthermore, during the life of the female, the mammary gland can undergo many rounds of expansion and proliferation. The mammary gland thus provides an excellent model for studying the ‘stem/progenitor’ cells that allow this repeated expansion and renewal. In this Review, we provide an overview of the different cell types that constitute the mammary gland, and discuss how these cell types arise and differentiate. As cellular differentiation cannot occur without proper signals, we also describe how the tissue microenvironment influences mammary gland development.
Collapse
Affiliation(s)
- Jamie L. Inman
- Life Sciences Division, Lawrence Berkeley National Laboratory, University of California, Berkeley, CA 94720, USA
| | - Claire Robertson
- Life Sciences Division, Lawrence Berkeley National Laboratory, University of California, Berkeley, CA 94720, USA
| | - Joni D. Mott
- Life Sciences Division, Lawrence Berkeley National Laboratory, University of California, Berkeley, CA 94720, USA
| | - Mina J. Bissell
- Life Sciences Division, Lawrence Berkeley National Laboratory, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
148
|
Brisken C, Ataca D. Endocrine hormones and local signals during the development of the mouse mammary gland. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 4:181-95. [PMID: 25645332 DOI: 10.1002/wdev.172] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 12/02/2014] [Accepted: 12/08/2014] [Indexed: 01/03/2023]
Abstract
Most of mammary gland development occurs postnatally under the control of female reproductive hormones, which in turn interact with other endocrine factors. While hormones impinge on many tissues and trigger very complex biological responses, tissue recombination experiments with hormone receptor-deficient mammary epithelia revealed eminent roles for estrogens, progesterone, and prolactin receptor (PrlR) signaling that are intrinsic to the mammary epithelium. A subset of the luminal mammary epithelial cells expresses the estrogen receptor α (ERα), the progesterone receptor (PR), and the PrlR and act as sensor cells. These cells convert the detected systemic signals into local signals that are developmental stage-dependent and may be direct, juxtacrine, or paracrine. This setup ensures that the original input is amplified and that the biological responses of multiple cell types can be coordinated. Some key mediators of hormone action have been identified such as Wnt, EGFR, IGFR, and RANK signaling. Multiple signaling pathways such as FGF, Hedgehog, and Notch signaling participate in driving different aspects of mammary gland development locally but how they link to the hormonal control remains to be elucidated. An increasing number of endocrine factors are appearing to have a role in mammary gland development, the adipose tissue is increasingly recognized to play a role in endocrine regulation, and a complex role of the immune system with multiple different cell types is being revealed. For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Cathrin Brisken
- ISREC - Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | |
Collapse
|
149
|
|
150
|
Mucosal Eosinophils. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00044-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|