1
|
Weinhaus B, Homan S, Kincaid M, Tadwalkar A, Gu X, Kumar S, Slaughter A, Zhang J, Wu Q, Kofron JM, Troutman TD, DeFalco T, Lucas D. Differential regulation of fetal bone marrow and liver hematopoiesis by yolk-sac-derived myeloid cells. Nat Commun 2025; 16:4427. [PMID: 40368906 DOI: 10.1038/s41467-025-59058-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 04/10/2025] [Indexed: 05/16/2025] Open
Abstract
Fetal hematopoiesis takes place in the liver before colonizing the bone marrow where it will persist for life. This colonization is thought to be mediated by specification of a microenvironment that selectively recruits hematopoietic cells to the nascent bone marrow. The identity and mechanisms regulating the specification of this colonization niche are unclear. Here we identify a VCAM1+ sinusoidal colonization niche in the diaphysis that regulates neutrophil and hematopoietic stem cell colonization of the bone marrow. Using confocal microscopy, we find that colonizing hematopoietic stem and progenitor cells (HSPC) and myeloid cells selectively localize to a subset of VCAM1+ sinusoids in the center of the diaphysis. Vcam1 deletion in endothelial cells impairs hematopoietic colonization while depletion of yolk-sac-derived osteoclasts disrupts VCAM1+ expression, and impairs neutrophil and HSPC colonization to the bone marrow. Depletion of yolk-sac-derived myeloid cells increases fetal liver hematopoietic stem cell numbers, function and erythropoiesis independent of osteoclast activity. Thus, the yolk sac produces myeloid cells that have opposite roles in fetal hematopoiesis: while yolk-sac derived myeloid cells in the bone marrow promote hematopoietic colonization by specifying a VCAM1+ colonization niche, a different subset of yolk-sac-derived myeloid cells inhibits HSC in the fetal liver.
Collapse
Affiliation(s)
- Benjamin Weinhaus
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Shelli Homan
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
| | - Morgan Kincaid
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
| | - Aryan Tadwalkar
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
| | - Xiaowei Gu
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Sumit Kumar
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
| | - Anastasiya Slaughter
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Jizhou Zhang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Qingqing Wu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - J Matthew Kofron
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Ty D Troutman
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Tony DeFalco
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Daniel Lucas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
2
|
Wang Z, Wang M, Liu J, Zhao D, Wang J, Wei F. Macrophage is crucial for tongue development by regulating myogenesis and vascularization. BMC Oral Health 2025; 25:678. [PMID: 40316997 PMCID: PMC12049047 DOI: 10.1186/s12903-025-06059-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 04/24/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND Abnormal tongue development is a craniofacial deformity that affects dental-maxillofacial esthetics and function. Recent evidence has identified macrophages as multi-functional immune cells crucial for heart and brain development. However, it is still unknown whether macrophages affect tongue development. Therefore, this study aims to assess the distribution, phenotype, and roles of macrophages in the developing tongue. METHODS In this study, immunohistochemical (IHC) and multiplex immunofluorescence (mIF) staining were conducted on C57BL/6 mice at embryonic day (E) 13.5, E14.5, E16.5, and E18.5 to analyze the distribution and phenotype of macrophages. Hematoxylin-Eosin (HE), IHC, IF, and mIF staining were also performed on embryonic CX3 CR1-CreERT2; Rosa-DTA conditional macrophage-depleted mice to investigate the effects on fetal tongue development and elucidate mechanisms from myogenesis, vascularization, and cell apoptosis. Statistical significance between the two groups was determined using unpaired two-tailed Student's t-tests. For comparisons involving three or more groups, one-way analysis of variance (ANOVA) followed by Tukey's multiple comparison tests was utilized. A significance level of P < 0.05 was set for statistical significance. RESULTS Macrophages were present in the developing tongue from E13.5 to E18.5, with a majority being of the M2 phenotype. Depletion of macrophages resulted in abnormal tongue morphology, decreased tongue height, width, and size, as well as reduced and disorganized muscle fibers. Depletion of macrophages also increased apoptosis. Vascular morphogenesis was impacted, with reductions in the luminal and vascular wall cross-sectional areas of the lingual artery. Vascular endothelial cells were reduced and disorganized with decreased expression of VEGFA and TGF-β1. Moreover, macrophages were located near vascular endothelial cells and secreted pro-angiogenic factors, suggesting their involvement in promoting vascularization. CONCLUSIONS Our findings indicate that macrophages play crucial roles in fetal tongue development by affecting myogenesis, cell apoptosis, and vascular growth.
Collapse
Affiliation(s)
- Ziyao Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, Jinan, Shandong, 250012, China
| | - Mengqiao Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, Jinan, Shandong, 250012, China
| | - Jiani Liu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, Jinan, Shandong, 250012, China
| | - Delu Zhao
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, Jinan, Shandong, 250012, China
| | - Jixiao Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, Jinan, Shandong, 250012, China
| | - Fulan Wei
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, Jinan, Shandong, 250012, China.
| |
Collapse
|
3
|
Lehtonen H, Jokela H, Hofmann J, Tola L, Mehmood A, Ginhoux F, Becher B, Greter M, Yegutkin GG, Salmi M, Gerke H, Rantakari P. Early precursor-derived pituitary gland tissue-resident macrophages play a pivotal role in modulating hormonal balance. Cell Rep 2025; 44:115227. [PMID: 39841599 DOI: 10.1016/j.celrep.2024.115227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 11/26/2024] [Accepted: 12/30/2024] [Indexed: 01/24/2025] Open
Abstract
The pituitary gland is the central endocrine regulatory organ producing and releasing hormones that coordinate major body functions. The physical location of the pituitary gland at the base of the brain, though outside the protective blood-brain barrier, leads to an unexplored special immune environment. Using single-cell transcriptomics, fate mapping, and imaging, we characterize pituitary-resident macrophages (pitMØs), revealing their heterogeneity and spatial specialization. Microglia-like macrophages (ml-MACs) are enriched in the posterior pituitary, while other pitMØs in the anterior pituitary exhibit close interactions with hormone-secreting cells. Importantly, all pitMØs originate from early yolk sac progenitors and maintain themselves through self-renewal, independent of bone marrow-derived monocytes. Macrophage depletion experiments unveil the role of macrophages in regulating intrapituitary hormonal balance through extracellular ATP-mediated intercellular signaling. Altogether, these findings provide information on pituitary gland macrophages and advance our understanding of immune-endocrine system crosstalk.
Collapse
Affiliation(s)
- Henna Lehtonen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Institute of Biomedicine, University of Turku, 20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | - Heli Jokela
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Institute of Biomedicine, University of Turku, 20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | - Julian Hofmann
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Institute of Biomedicine, University of Turku, 20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | - Lauriina Tola
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Institute of Biomedicine, University of Turku, 20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | - Arfa Mehmood
- Institute of Biomedicine, University of Turku, 20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | - Florent Ginhoux
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648, Singapore; INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France; Translational Immunology Institute, SingHealth Duke-NUS, Singapore 169856, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland
| | - Gennady G Yegutkin
- InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland; MediCity Research Laboratory, University of Turku, 20520 Turku, Finland
| | - Marko Salmi
- Institute of Biomedicine, University of Turku, 20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland; MediCity Research Laboratory, University of Turku, 20520 Turku, Finland
| | - Heidi Gerke
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Institute of Biomedicine, University of Turku, 20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | - Pia Rantakari
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Institute of Biomedicine, University of Turku, 20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland.
| |
Collapse
|
4
|
Murrey MW, Ng IT, Pixley FJ. The role of macrophage migratory behavior in development, homeostasis and tumor invasion. Front Immunol 2024; 15:1480084. [PMID: 39588367 PMCID: PMC11586339 DOI: 10.3389/fimmu.2024.1480084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/23/2024] [Indexed: 11/27/2024] Open
Abstract
Tumor-associated macrophages (TAMs) recapitulate the developmental and homeostatic behaviors of tissue resident macrophages (TRMs) to promote tumor growth, invasion and metastasis. TRMs arise in the embryo and colonize developing tissues, initially to guide tissue morphogenesis and then to form complex networks in adult tissues to constantly search for threats to homeostasis. The macrophage growth factor, colony-stimulating factor-1 (CSF-1), which is essential for TRM survival and differentiation, is also responsible for the development of the unique motility machinery of mature macrophages that underpins their ramified morphologies, migratory capacity and ability to degrade matrix. Two CSF-1-activated kinases, hematopoietic cell kinase and the p110δ catalytic isoform of phosphatidylinositol 3-kinase, regulate this machinery and selective inhibitors of these proteins completely block macrophage invasion. Considering tumors co-opt the invasive capacity of TAMs to promote their own invasion, these proteins are attractive targets for drug development to inhibit tumor progression to invasion and metastasis.
Collapse
Affiliation(s)
| | | | - Fiona J. Pixley
- Macrophage Biology and Cancer Laboratory, School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
5
|
Friedman-DeLuca M, Karagiannis GS, Condeelis JS, Oktay MH, Entenberg D. Macrophages in tumor cell migration and metastasis. Front Immunol 2024; 15:1494462. [PMID: 39555068 PMCID: PMC11563815 DOI: 10.3389/fimmu.2024.1494462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/15/2024] [Indexed: 11/19/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are a phenotypically diverse, highly plastic population of cells in the tumor microenvironment (TME) that have long been known to promote cancer progression. In this review, we summarize TAM ontogeny and polarization, and then explore how TAMs enhance tumor cell migration through the TME, thus facilitating metastasis. We also discuss how chemotherapy and host factors including diet, obesity, and race, impact TAM phenotype and cancer progression. In brief, TAMs induce epithelial-mesenchymal transition (EMT) in tumor cells, giving them a migratory phenotype. They promote extracellular matrix (ECM) remodeling, allowing tumor cells to migrate more easily. TAMs also provide chemotactic signals that promote tumor cell directional migration towards blood vessels, and then participate in the signaling cascade at the blood vessel that allows tumor cells to intravasate and disseminate throughout the body. Furthermore, while chemotherapy can repolarize TAMs to induce an anti-tumor response, these cytotoxic drugs can also lead to macrophage-mediated tumor relapse and metastasis. Patient response to chemotherapy may be dependent on patient-specific factors such as diet, obesity, and race, as these factors have been shown to alter macrophage phenotype and affect cancer-related outcomes. More research on how chemotherapy and patient-specific factors impact TAMs and cancer progression is needed to refine treatment strategies for cancer patients.
Collapse
Affiliation(s)
- Madeline Friedman-DeLuca
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - George S. Karagiannis
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Microbiology and Immunology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Marilyn and Stanley M. Katz Institute for Immunotherapy of Cancer and Inflammatory Disorders, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - John S. Condeelis
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Cell Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - Maja H. Oktay
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - David Entenberg
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| |
Collapse
|
6
|
Chi J, Gao Q, Liu D. Tissue-Resident Macrophages in Cancer: Friend or Foe? Cancer Med 2024; 13:e70387. [PMID: 39494816 PMCID: PMC11533131 DOI: 10.1002/cam4.70387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/15/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024] Open
Abstract
INTRODUCTION Macrophages are essential in maintaining homeostasis, combating infections, and influencing the process of various diseases, including cancer. Macrophages originate from diverse lineages: Notably, tissue-resident macrophages (TRMs) differ from hematopoietic stem cells and circulating monocyte-derived macrophages based on genetics, development, and function. Therefore, understanding the recruited and TRM populations is crucial for investigating disease processes. METHODS By searching literature databses, we summarized recent relevant studies. Research has shown that tumor-associated macrophages (TAMs) of distinct origins accumulate in tumor microenvironment (TME), with TRM-derived TAMs closely resembling gene signatures of normal TRMs. RESULTS Recent studies have revealed that TRMs play a crucial role in cancer progression. However, organ-specific effects complicate TRM investigations. Nonetheless, the precise involvement of TRMs in tumors is unclear. This review explores the multifaceted roles of TRMs in cancer, presenting insights into their origins, proliferation, the latest research methodologies, their impact across various tumor sites, their potential and strategies as therapeutic targets, interactions with other cells within the TME, and the internal heterogeneity of TRMs. CONCLUSIONS We believe that a comprehensive understanding of the multifaceted roles of TRMs will pave the way for targeted TRM therapies in the treatment of cancer.
Collapse
Affiliation(s)
- Jianhua Chi
- Department of Obstetrics and GynecologyNational Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and MetastasisTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Qinglei Gao
- Department of Obstetrics and GynecologyNational Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and MetastasisTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Dan Liu
- Department of Obstetrics and GynecologyNational Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and MetastasisTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
7
|
Feng SW, North TM, Wivell P, Pletcher A, Popratiloff A, Shibata M. Macrophages of multiple hematopoietic origins reside in the developing prostate. Development 2024; 151:dev203070. [PMID: 39082371 PMCID: PMC11385323 DOI: 10.1242/dev.203070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024]
Abstract
Tissue-resident macrophages contribute to the organogenesis of many tissues. Growth of the prostate is regulated by androgens during puberty, yet androgens are considered immune suppressive. In this study, we characterized the localization, androgen receptor expression and hematopoietic origin of prostate macrophages, and transiently ablated macrophages during postnatal prostate organogenesis in the mouse. We show that myeloid cells were abundant in the prostate during puberty. However, nuclear androgen receptor expression was not detected in most macrophages. We found Cx3cr1, a marker for macrophages, monocytes and dendritic cells, expressed in interstitial macrophages surrounding the prostate and associated with nerve fibers. Furthermore, we provide evidence for the co-existence of embryonic origin, self-renewing, tissue-resident macrophages and recruited macrophages of bone-marrow monocyte origin in the prostate during puberty. Our findings suggest that prostate macrophages promote neural patterning and may shed further light on our understanding of the role of the innate immune system in prostate pathology in response to inflammation and in cancer.
Collapse
Affiliation(s)
- Sally W Feng
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
- The George Washington University Cancer Center, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Tanya M North
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
- The George Washington University Cancer Center, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Peri Wivell
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
- The George Washington University Cancer Center, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Andrew Pletcher
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
- The George Washington University Cancer Center, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Anastas Popratiloff
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
- GW Nanofabrication and Imaging Center, The George Washington University, Washington, DC 20052, USA
| | - Maho Shibata
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
- The George Washington University Cancer Center, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| |
Collapse
|
8
|
Biswas M. Understanding tissue-resident macrophages unlocks the potential for novel combinatorial strategies in breast cancer. Front Immunol 2024; 15:1375528. [PMID: 39104525 PMCID: PMC11298421 DOI: 10.3389/fimmu.2024.1375528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/14/2024] [Indexed: 08/07/2024] Open
Abstract
Tissue-resident macrophages (TRMs) are an integral part of the innate immune system, but their biology is not well understood in the context of cancer. Distinctive resident macrophage populations are identified in different organs in mice using fate mapping studies. They develop from the yolk sac and self-maintain themselves lifelong in specific tissular niches. Similarly, breast-resident macrophages are part of the mammary gland microenvironment. They reside in the breast adipose tissue stroma and close to the ductal epithelium and help in morphogenesis. In breast cancer, TRMs may promote disease progression and metastasis; however, precise mechanisms have not been elucidated. TRMs interact intimately with recruited macrophages, cytotoxic T cells, and other immune cells along with cancer cells, deciding further immunosuppressive or cytotoxic pathways. Moreover, triple-negative breast cancer (TNBC), which is generally associated with poor outcomes, can harbor specific TRM phenotypes. The influence of TRMs on adipose tissue stroma of the mammary gland also contributes to tumor progression. The complex crosstalk between TRMs with T cells, stroma, and breast cancer cells can establish a cascade of downstream events, understanding which can offer new insight for drug discovery and upcoming treatment choices. This review aims to acknowledge the previous research done in this regard while exploring existing research gaps and the future therapeutic potential of TRMs as a combination or single agent in breast cancer.
Collapse
Affiliation(s)
- Manjusha Biswas
- Department of Molecular Biomedicine, Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| |
Collapse
|
9
|
Ammarah U, Pereira‐Nunes A, Delfini M, Mazzone M. From monocyte-derived macrophages to resident macrophages-how metabolism leads their way in cancer. Mol Oncol 2024; 18:1739-1758. [PMID: 38411356 PMCID: PMC11223613 DOI: 10.1002/1878-0261.13618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/24/2024] [Accepted: 02/16/2024] [Indexed: 02/28/2024] Open
Abstract
Macrophages are innate immune cells that play key roles during both homeostasis and disease. Depending on the microenvironmental cues sensed in different tissues, macrophages are known to acquire specific phenotypes and exhibit unique features that, ultimately, orchestrate tissue homeostasis, defense, and repair. Within the tumor microenvironment, macrophages are referred to as tumor-associated macrophages (TAMs) and constitute a heterogeneous population. Like their tissue resident counterpart, TAMs are plastic and can switch function and phenotype according to the niche-derived stimuli sensed. While changes in TAM phenotype are known to be accompanied by adaptive alterations in their cell metabolism, it is reported that metabolic reprogramming of macrophages can dictate their activation state and function. In line with these observations, recent research efforts have been focused on defining the metabolic traits of TAM subsets in different tumor malignancies and understanding their role in cancer progression and metastasis formation. This knowledge will pave the way to novel therapeutic strategies tailored to cancer subtype-specific metabolic landscapes. This review outlines the metabolic characteristics of distinct TAM subsets and their implications in tumorigenesis across multiple cancer types.
Collapse
Affiliation(s)
- Ummi Ammarah
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer BiologyVIBLeuvenBelgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer BiologyKU LeuvenBelgium
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CentreUniversity of TorinoItaly
| | - Andreia Pereira‐Nunes
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer BiologyVIBLeuvenBelgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer BiologyKU LeuvenBelgium
- Life and Health Sciences Research Institute (ICVS), School of MedicineUniversity of MinhoBragaPortugal
- ICVS/3B's‐PT Government Associate LaboratoryBraga/GuimarãesPortugal
| | - Marcello Delfini
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer BiologyVIBLeuvenBelgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer BiologyKU LeuvenBelgium
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer BiologyVIBLeuvenBelgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer BiologyKU LeuvenBelgium
| |
Collapse
|
10
|
Elfstrum AK, Bapat AS, Schwertfeger KL. Defining and targeting macrophage heterogeneity in the mammary gland and breast cancer. Cancer Med 2024; 13:e7053. [PMID: 38426622 PMCID: PMC10905685 DOI: 10.1002/cam4.7053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/09/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024] Open
Abstract
INTRODUCTION Macrophages are innate immune cells that are associated with extensive phenotypic and functional plasticity and contribute to normal development, tissue homeostasis, and diseases such as cancer. In this review, we discuss the heterogeneity of tissue resident macrophages in the normal mammary gland and tumor-associated macrophages in breast cancer. Tissue resident macrophages are required for mammary gland development, where they have been implicated in promoting extracellular matrix remodeling, apoptotic clearance, and cellular crosstalk. In the context of cancer, tumor-associated macrophages are key drivers of growth and metastasis via their ability to promote matrix remodeling, angiogenesis, lymphangiogenesis, and immunosuppression. METHOD We identified and summarized studies in Pubmed that describe the phenotypic and functional heterogeneity of macrophages and the implications of targeting individual subsets, specifically in the context of mammary gland development and breast cancer. We also identified and summarized recent studies using single-cell RNA sequencing to identify and describe macrophage subsets in human breast cancer samples. RESULTS Advances in single-cell RNA sequencing technologies have yielded nuances in macrophage heterogeneity, with numerous macrophage subsets identified in both the normal mammary gland and breast cancer tissue. Macrophage subsets contribute to mammary gland development and breast cancer progression in differing ways, and emerging studies highlight a role for spatial localization in modulating their phenotype and function. CONCLUSION Understanding macrophage heterogeneity and the unique functions of each subset in both normal mammary gland development and breast cancer progression may lead to more promising targets for the treatment of breast cancer.
Collapse
Affiliation(s)
- Alexis K. Elfstrum
- Microbiology, Immunology, and Cancer Biology Graduate ProgramUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Aditi S. Bapat
- Molecular Pharmacology and Therapeutics Graduate ProgramUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Kathryn L. Schwertfeger
- Department of Laboratory Medicine and PathologyUniversity of MinnesotaMinneapolisMinnesotaUSA
- Masonic Cancer CenterUniversity of MinnesotaMinneapolisMinnesotaUSA
- Center for ImmunologyUniversity of MinnesotaMinneapolisMinnesotaUSA
| |
Collapse
|
11
|
Yang X, Xu H, Yang X, Wang H, Zou L, Yang Q, Qi X, Li L, Duan H, Yan X, Fu NY, Tan J, Hou Z, Jiao B. Mcam inhibits macrophage-mediated development of mammary gland through non-canonical Wnt signaling. Nat Commun 2024; 15:36. [PMID: 38167296 PMCID: PMC10761817 DOI: 10.1038/s41467-023-44338-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
While canonical Wnt signaling is well recognized for its crucial regulatory functions in cell fate decisions, the role of non-canonical Wnt signaling in adult stem cells remains elusive and contradictory. Here, we identified Mcam, a potential member of the non-canonical Wnt signaling, as an important negative regulator of mammary gland epithelial cells (MECs) by genome-scale CRISPR-Cas9 knockout (GeCKO) library screening. Loss of Mcam increases the clonogenicity and regenerative capacity of MECs, and promotes the proliferation, differentiation, and ductal morphogenesis of mammary epithelial in knockout mice. Mechanically, Mcam knockout recruits and polarizes macrophages through the Il4-Stat6 axis, thereby promoting secretion of the non-canonical Wnt ligand Wnt5a and its binding to the non-canonical Wnt signaling receptor Ryk to induce the above phenotypes. These findings reveal Mcam roles in mammary gland development by orchestrating communications between MECs and macrophages via a Wnt5a/Ryk axis, providing evidences for non-canonical Wnt signaling in mammary development.
Collapse
Affiliation(s)
- Xing Yang
- Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, 650051, China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, 650051, China
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Chinese Academy of Sciences, Kunming, Yunnan, 650201, China
| | - Haibo Xu
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Chinese Academy of Sciences, Kunming, Yunnan, 650201, China
| | - Xu Yang
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Chinese Academy of Sciences, Kunming, Yunnan, 650201, China
| | - Hui Wang
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Chinese Academy of Sciences, Kunming, Yunnan, 650201, China
| | - Li Zou
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Chinese Academy of Sciences, Kunming, Yunnan, 650201, China
| | - Qin Yang
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Chinese Academy of Sciences, Kunming, Yunnan, 650201, China
| | - Xiaopeng Qi
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Chinese Academy of Sciences, Kunming, Yunnan, 650201, China
| | - Li Li
- Research Center of Stem cells and Ageing, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Hongxia Duan
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100000, China
| | - Xiyun Yan
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100000, China
| | - Nai Yang Fu
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, 169857, Singapore
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Jing Tan
- Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, 650051, China.
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, 650051, China.
| | - Zongliu Hou
- Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, 650051, China.
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, 650051, China.
| | - Baowei Jiao
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Chinese Academy of Sciences, Kunming, Yunnan, 650201, China.
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650201, China.
| |
Collapse
|
12
|
Ahlback A, Gentek R. Fate-Mapping Macrophages: From Ontogeny to Functions. Methods Mol Biol 2024; 2713:11-43. [PMID: 37639113 DOI: 10.1007/978-1-0716-3437-0_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
Macrophages are vital to the physiological function of most tissues, but also contribute to disease through a multitude of pathological roles. They are thus highly plastic and heterogeneous. It is now well recognized that macrophages develop from several distinct progenitors from embryogenesis onwards and extending throughout life. Tissue-resident macrophages largely originate from embryonic sources and in many cases self-maintain independently without monocyte input. However, in certain tissues, monocyte-derived macrophages replace these over time or as a result of tissue injury and inflammation. This additional layer of heterogeneity has introduced many questions regarding the influence of origin on fate and function of macrophages in health and disease. To comprehensively address these questions, appropriate methods of tracing macrophage ontogeny are required. This chapter explores why ontogeny is of vital importance in macrophage biology and how to delineate macrophage populations by origin through genetic fate mapping. First, we summarize the current view of macrophage ontogeny and briefly discuss how origin may influence macrophage function in homeostasis and pathology. We go on to make the case for genetic fate mapping as the gold standard and briefly review different fate-mapping models. We then put forward our recommendations for fate-mapping strategies best suited to answer specific research questions and finally discuss the strengths and limitations of currently available models.
Collapse
Affiliation(s)
- Anna Ahlback
- The University of Edinburgh, Institute for Regeneration and Repair, Centre for Reproductive Health & Centre for Inflammation Research, Edinburgh, UK
| | - Rebecca Gentek
- The University of Edinburgh, Institute for Regeneration and Repair, Centre for Reproductive Health & Centre for Inflammation Research, Edinburgh, UK.
| |
Collapse
|
13
|
Boutas I, Kontogeorgi A, Kalantaridou SN, Dimitrakakis C, Patsios P, Kalantzi M, Xanthos T. Reverse Onco-Cardiology: What Is the Evidence for Breast Cancer? A Systematic Review of the Literature. Int J Mol Sci 2023; 24:16500. [PMID: 38003690 PMCID: PMC10671526 DOI: 10.3390/ijms242216500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/12/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Breast cancer and cardiovascular diseases (CVD) represent significant global health challenges, with CVD being the leading cause of mortality and breast cancer, showing a complex pattern of incidence and mortality. We explore the intricate interplay between these two seemingly distinct medical conditions, shedding light on their shared risk factors and potential pathophysiological connections. A specific connection between hypertension (HTN), atrial fibrillation (AF), myocardial infarction (MI), and breast cancer was evaluated. HTN is explored in detail, emphasizing the role of aging, menopause, insulin resistance, and obesity as common factors linking HTN and breast cancer. Moreover, an attempt is made to identify the potential impact of antihypertensive medications and highlight the increased risk of breast cancer among those women, with a focus on potential mechanisms. A summary of key findings underscores the need for a multisystem approach to understanding the relationship between CVD and breast cancer is also explored with a highlight for all the gaps in current research, such as the lack of clinical observational data on MI and breast cancer in humans and the need for studies specifically designed for breast cancer. This paper concludes that there should be a focus on potential clinical applications of further investigation in this field, including personalized prevention and screening strategies for women at risk. Overall, the authors attempt to provide a comprehensive overview of the intricate connections between breast cancer and cardiovascular diseases, emphasizing the importance of further research in this evolving field of cardio-oncology.
Collapse
Affiliation(s)
- Ioannis Boutas
- Breast Unit, Rea Maternity Hospital, 383 Andrea Siggrou Ave., Paleo Faliro, 175 64 Athens, Greece
| | - Adamantia Kontogeorgi
- Medical School, University of Crete, 13 Andrea Kalokairinoy Ave., 715 00 Giofirakia, Greece
| | - Sophia N. Kalantaridou
- Third Department of Obstetrics and Gynecology, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 1 Rimini Str., 124 62 Chaidari, Greece;
| | - Constantine Dimitrakakis
- First Department of Obstetrics and Gynecology, Alexandra University Hospital, Medical School, National and Kapodistrian University of Athens, 4-2 Lourou Str., 115 28 Athens, Greece;
| | - Panagiotis Patsios
- Cardiology Department, Elpis General Hospital, 7 Dimitsana Str., 115 22 Athens, Greece;
| | - Maria Kalantzi
- Post Graduate Study Program “Cardiopulmonary Resuscitation”, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 115 27 Athens, Greece;
| | - Theodoros Xanthos
- School of Health Sciences, University of West Attica, 28 Aghiou Spyridonos Str., 122 43 Aigaleo, Greece;
| |
Collapse
|
14
|
Li X, Jiang F, Hu Y, Lang Z, Zhan Y, Zhang R, Tao Q, Luo C, Yu J, Zheng J. Schisandrin B Promotes Hepatic Stellate Cell Ferroptosis via Wnt Pathway-Mediated Ly6C lo Macrophages. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37922022 DOI: 10.1021/acs.jafc.3c03409] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2023]
Abstract
A key event in liver fibrosis is the activation of the hepatic stellate cell (HSC). Schisandrin B (Sch B), a major component extracted from Schisandra chinensis, has been shown to inhibit HSC activation. Recently, ferroptosis (FPT) has been reported to be involved in HSC activation. However, whether Sch B has an effect on the HSC FPT remains unclear. Herein, we explored the effects of Sch B on liver fibrosis in vivo and in vitro and the roles of Wnt agonist 1 and ferrostatin-1 in the antifibrotic effects of Sch B. Sch B effectively alleviated CCl4-induced liver fibrosis, with decreased collagen deposition and α-SMA level. Additionally, Sch B resulted in an increase in lymphocyte antigen 6 complex locus C low (Ly6Clo) macrophages, contributing to a reduced level of TIMP1 and increased MMP2. Notably, the Wnt pathway was involved in Sch B-mediated Ly6C macrophage phenotypic transformation. Further studies demonstrated that Sch B-treated macrophages had an inhibitory effect on HSC activation, which was associated with HSC FPT. GPX4, a negative regulator of FPT, was induced by Sch B and found to be involved in the crosstalk between macrophage and HSC FPT. Furthermore, HSC inactivation as well as FPT induced by Sch B-treated macrophages was blocked down by Wnt pathway agonist 1. Collectively, we demonstrate that Sch B inhibits liver fibrosis, at least partially, through mediating Ly6Clo macrophages and HSC FPT. Sch B enhances Wnt pathway inactivation, leading to the increase in Ly6Clo macrophages, which contributes to HSC FPT. Sch B may be a promising drug for liver fibrosis treatment.
Collapse
Affiliation(s)
- Xinmiao Li
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Feng Jiang
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yuhang Hu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Zhichao Lang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yating Zhan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Rongrong Zhang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Qiqi Tao
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Chengchu Luo
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Jinglu Yu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Department of Laboratory Medicine, Lishui Municipal Central Hospital,Lishui 323020, China
| | - Jianjian Zheng
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
15
|
Nahmias-Blank D, Maimon O, Meirovitz A, Sheva K, Peretz-Yablonski T, Elkin M. Excess body weight and postmenopausal breast cancer: Emerging molecular mechanisms and perspectives. Semin Cancer Biol 2023; 96:26-35. [PMID: 37739109 DOI: 10.1016/j.semcancer.2023.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023]
Abstract
Postmenopausal, obese women have a significantly higher risk of developing estrogen receptor-positive (ER+) breast tumors, that are resistant to therapies and are associated with higher recurrence and death rates. The global prevalence of overweight/obese women has reached alarming proportions and with postmenopausal ER+ breast carcinoma (BC) having the highest incidence among the three obesity-related cancers in females (i.e., breast, endometrial and ovarian), this is of significant concern. Elucidation of the precise molecular mechanisms underlying the pro-cancerous action of obesity in ER+BC is therefore critical for disease prevention and novel treatment initiatives. Interestingly, accumulating data has shown opposing relationships between obesity and cancer in either pre- or post-menopausal women. Excess body weight is associated with an increased risk of breast cancer in postmenopausal women and a decreased risk in pre-menopausal women. Moreover, excess adiposity during early life appears to be protective against postmenopausal breast cancer, including both ER+ and ER negative BC subtypes. Overall, estrogen-dependent mechanisms have been implicated as the main driving force in obesity-related breast tumorigenesis. In the present review we discuss the epidemiologic and mechanistic aspects of association between obesity and breast tumors after menopause, mainly in the context of hormone dependency. Molecular and cellular events underlying this association present as potential avenues for both therapeutic intervention as well as the prevention of BC-promoting processes linked to excess adiposity, which is proving to be vital in an increasingly obese global population.
Collapse
Affiliation(s)
- Daniela Nahmias-Blank
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ofra Maimon
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Amichay Meirovitz
- Legacy Heritage Oncology Center and Dr. Larry Norton Institute, Soroka University Medical Center, Be'er Sheva 84101, Israel
| | - Kim Sheva
- Legacy Heritage Oncology Center and Dr. Larry Norton Institute, Soroka University Medical Center, Be'er Sheva 84101, Israel
| | - Tamar Peretz-Yablonski
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel; Hebrew University Medical School, Jerusalem 91120, Israel
| | - Michael Elkin
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel; Hebrew University Medical School, Jerusalem 91120, Israel.
| |
Collapse
|
16
|
Ben-Chetrit N, Niu X, Sotelo J, Swett AD, Rajasekhar VK, Jiao MS, Stewart CM, Bhardwaj P, Kottapalli S, Ganesan S, Loyher PL, Potenski C, Hannuna A, Brown KA, Iyengar NM, Giri DD, Lowe SW, Healey JH, Geissmann F, Sagi I, Joyce JA, Landau DA. Breast Cancer Macrophage Heterogeneity and Self-renewal are Determined by Spatial Localization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563749. [PMID: 37961223 PMCID: PMC10634790 DOI: 10.1101/2023.10.24.563749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Tumor-infiltrating macrophages support critical steps in tumor progression, and their accumulation in the tumor microenvironment (TME) is associated with adverse outcomes and therapeutic resistance across human cancers. In the TME, macrophages adopt diverse phenotypic alterations, giving rise to heterogeneous immune activation states and induction of cell cycle. While the transcriptional profiles of these activation states are well-annotated across human cancers, the underlying signals that regulate macrophage heterogeneity and accumulation remain incompletely understood. Here, we leveraged a novel ex vivo organotypic TME (oTME) model of breast cancer, in vivo murine models, and human samples to map the determinants of functional heterogeneity of TME macrophages. We identified a subset of F4/80highSca-1+ self-renewing macrophages maintained by type-I interferon (IFN) signaling and requiring physical contact with cancer-associated fibroblasts. We discovered that the contact-dependent self-renewal of TME macrophages is mediated via Notch4, and its inhibition abrogated tumor growth of breast and ovarian carcinomas in vivo, as well as lung dissemination in a PDX model of triple-negative breast cancer (TNBC). Through spatial multi-omic profiling of protein markers and transcriptomes, we found that the localization of macrophages further dictates functionally distinct but reversible phenotypes, regardless of their ontogeny. Whereas immune-stimulatory macrophages (CD11C+CD86+) populated the tumor epithelial nests, the stroma-associated macrophages (SAMs) were proliferative, immunosuppressive (Sca-1+CD206+PD-L1+), resistant to CSF-1R depletion, and associated with worse patient outcomes. Notably, following cessation of CSF-1R depletion, macrophages rebounded primarily to the SAM phenotype, which was associated with accelerated growth of mammary tumors. Our work reveals the spatial determinants of macrophage heterogeneity in breast cancer and highlights the disruption of macrophage self-renewal as a potential new therapeutic strategy.
Collapse
Affiliation(s)
- Nir Ben-Chetrit
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
- These authors contributed equally
| | - Xiang Niu
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
- These authors contributed equally
- Present address: Genentech, Inc., South San Francisco, CA, USA
| | - Jesus Sotelo
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Ariel D. Swett
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Vinagolu K. Rajasekhar
- Orthopedic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria S. Jiao
- Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Caitlin M. Stewart
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Priya Bhardwaj
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Sanjay Kottapalli
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Saravanan Ganesan
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Pierre-Louis Loyher
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Catherine Potenski
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Assaf Hannuna
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Kristy A. Brown
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Neil M. Iyengar
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dilip D. Giri
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Scott W. Lowe
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - John H. Healey
- Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Frederic Geissmann
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Johanna A. Joyce
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Switzerland
| | - Dan A. Landau
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| |
Collapse
|
17
|
Symonds EKC, Black B, Brown A, Meredith I, Currie MJ, Hally KE, Danielson KM. Adipose derived stem cell extracellular vesicles modulate primary human macrophages to an anti-inflammatory phenotype in vitro. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e104. [PMID: 38939512 PMCID: PMC11080877 DOI: 10.1002/jex2.104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/11/2023] [Accepted: 07/12/2023] [Indexed: 06/29/2024]
Abstract
EVs released by adipose derived stem cells (ADSCs) have shown promise as a therapeutic for tissue repair because of their purported immune-regulatory properties. Extracellular vesicles (EVs) from ADSCs could be beneficial in improving graft retention rates for autologous fat grafting (AFG) post-mastectomy as, currently, grafted tissue rates are variable. Enriching grafted tissue with ADSC-EVs may improve retention rates by modulating macrophages resident within both the breast and lipoaspirate. We aimed to identify key macrophage phenotypes that are modulated by ADSC-EVs in vitro. ADSCs were isolated from lipoaspirates of women undergoing AFG and characterised by flow cytometry and differentiation potential. ADSC-EVs were isolated from culture media and characterised by tuneable resistive pulse sensing, transmission electron microscopy and Western blot. Primary monocyte-derived macrophages were polarized to an M1-like (GM-CSF, IFNγ), M2-like phenotype (M-CSF, IL-4) or maintained (M0-like; M-CSF) and ADSC-EVs were co-cultured with macrophages for 48 h. Flow cytometry and high-dimensional analysis clustered macrophages post co-culture. A manual gating strategy was generated to recapitulate these clusters and was applied to a repeat experimental run. Both runs were analysed to examine the prevalence of each cluster, representing a unique macrophage phenotype, with and without ADSC-EVs. Following the addition of ADSC-EVs, M0-like macrophages demonstrated a reciprocal shift of cell distribution from a cluster with a 'high inflammatory profile' (CD36+++CD206+++CD86+++; 16.5 ± 7.0%; p < 0.0001) to a cluster with a 'lower inflammatory profile' (CD36+CD206+CD86+; 35 ± 21.5%; p < 0.05). M1-like macrophages shifted from a cluster with a 'high inflammatory profile' (CD206++CD11b++CD36++CD163++; 26.1 ± 9.4%; p = 0.0024) to a 'lower inflammatory profile' (CD206+CD11b+CD36+CD163+; 72.8 ± 8.7%; p = 0.0007). There was no shift in M2-like clusters following ADSC-EV treatment. ADSC-EVs are complex regulators of macrophage phenotype that can shift macrophages away from a heightened pro-inflammatory state.
Collapse
Affiliation(s)
- Emma K. C. Symonds
- Department of Surgery and AnaesthesiaUniversity of Otago WellingtonWellingtonNew Zealand
| | - Bianca Black
- Department of Surgery and AnaesthesiaUniversity of Otago WellingtonWellingtonNew Zealand
| | - Alexander Brown
- Department of General SurgeryWellington Regional HospitalWellingtonNew Zealand
| | - Ineke Meredith
- Department of General SurgeryWellington Regional HospitalWellingtonNew Zealand
| | - Margaret J. Currie
- Mackenzie Cancer Research GroupUniversity of Otago ChristchurchChristchurchNew Zealand
| | - Kathryn E. Hally
- Department of Surgery and AnaesthesiaUniversity of Otago WellingtonWellingtonNew Zealand
| | - Kirsty M. Danielson
- Department of Surgery and AnaesthesiaUniversity of Otago WellingtonWellingtonNew Zealand
| |
Collapse
|
18
|
Cansever D, Petrova E, Krishnarajah S, Mussak C, Welsh CA, Mildenberger W, Mulder K, Kreiner V, Roussel E, Stifter SA, Andreadou M, Zwicky P, Jurado NP, Rehrauer H, Tan G, Liu Z, Blériot C, Ronchi F, Macpherson AJ, Ginhoux F, Natalucci G, Becher B, Greter M. Lactation-associated macrophages exist in murine mammary tissue and human milk. Nat Immunol 2023:10.1038/s41590-023-01530-0. [PMID: 37337103 PMCID: PMC10307629 DOI: 10.1038/s41590-023-01530-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 05/08/2023] [Indexed: 06/21/2023]
Abstract
Macrophages are involved in immune defense, organogenesis and tissue homeostasis. Macrophages contribute to the different phases of mammary gland remodeling during development, pregnancy and involution postlactation. Less is known about the dynamics of mammary gland macrophages in the lactation stage. Here, we describe a macrophage population present during lactation in mice. By multiparameter flow cytometry and single-cell RNA sequencing, we identified a lactation-induced CD11c+CX3CR1+Dectin-1+ macrophage population (liMac) that was distinct from the two resident F4/80hi and F4/80lo macrophage subsets present pregestationally. LiMacs were predominantly monocyte-derived and expanded by proliferation in situ concomitant with nursing. LiMacs developed independently of IL-34, but required CSF-1 signaling and were partly microbiota-dependent. Locally, they resided adjacent to the basal cells of the alveoli and extravasated into the milk. We found several macrophage subsets in human milk that resembled liMacs. Collectively, these findings reveal the emergence of unique macrophages in the mammary gland and milk during lactation.
Collapse
Affiliation(s)
- Dilay Cansever
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Roche, Basel, Switzerland
| | - Ekaterina Petrova
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Caroline Mussak
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Christina A Welsh
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Wiebke Mildenberger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Kevin Mulder
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Université Paris-Saclay, Ile-de-France, France
| | - Victor Kreiner
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Elsa Roussel
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sebastian A Stifter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Myrto Andreadou
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Pascale Zwicky
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Hubert Rehrauer
- Functional Genomics Center Zurich, ETH Zurich/University of Zurich, Zurich, Switzerland
| | - Ge Tan
- Functional Genomics Center Zurich, ETH Zurich/University of Zurich, Zurich, Switzerland
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Camille Blériot
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut Necker des Enfants Malades, CNRS, Paris, France
| | - Francesca Ronchi
- University Clinic for Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Andrew J Macpherson
- University Clinic for Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Florent Ginhoux
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Giancarlo Natalucci
- Larsson-Rosenquist Center for Neurodevelopment, Growth and Nutrition of the Newborn, Department of Neonatology, University Hospital Zurich, Zurich, Switzerland
- Newborn Research, Department of Neonatology, University Hospital Zurich, Zurich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
19
|
Timperi E, Romano E. Stromal circuits involving tumor-associated macrophages and cancer-associated fibroblasts. Front Immunol 2023; 14:1194642. [PMID: 37342322 PMCID: PMC10277481 DOI: 10.3389/fimmu.2023.1194642] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/19/2023] [Indexed: 06/22/2023] Open
Abstract
The tumor associated macrophages (TAM) represent one of most abundant subpopulations across several solid cancers and their number/frequency is associated with a poor clinical outcome. It has been clearly demonstrated that stromal cells, such as the cancer associated fibroblasts (CAFs), may orchestrate TAM recruitment, survival and reprogramming. Today, single cell-RNA sequencing (sc-RNA seq) technologies allowed a more granular knowledge about TAMs and CAFs phenotypical and functional programs. In this mini-review we discuss the recent discoveries in the sc-RNA seq field focusing on TAM and CAF identity and their crosstalk in the tumor microenvironment (TME) of solid cancers.
Collapse
Affiliation(s)
- Eleonora Timperi
- Department of Immunology, INSERM U932, Université Paris Sciences et Lettres (PSL) Research University, Institut Curie, Paris, France
| | - Emanuela Romano
- Department of Immunology, INSERM U932, Université Paris Sciences et Lettres (PSL) Research University, Institut Curie, Paris, France
- Department of Medical Oncology, Center for Cancer Immunotherapy, Institut Curie, Paris, France
| |
Collapse
|
20
|
Kuziel G, Moore BN, Arendt LM. Obesity and Fibrosis: Setting the Stage for Breast Cancer. Cancers (Basel) 2023; 15:cancers15112929. [PMID: 37296891 DOI: 10.3390/cancers15112929] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Obesity is a rising health concern and is linked to a worsened breast cancer prognosis. Tumor desmoplasia, which is characterized by elevated numbers of cancer-associated fibroblasts and the deposition of fibrillar collagens within the stroma, may contribute to the aggressive clinical behavior of breast cancer in obesity. A major component of the breast is adipose tissue, and fibrotic changes in adipose tissue due to obesity may contribute to breast cancer development and the biology of the resulting tumors. Adipose tissue fibrosis is a consequence of obesity that has multiple sources. Adipocytes and adipose-derived stromal cells secrete extracellular matrix composed of collagen family members and matricellular proteins that are altered by obesity. Adipose tissue also becomes a site of chronic, macrophage-driven inflammation. Macrophages exist as a diverse population within obese adipose tissue and mediate the development of fibrosis through the secretion of growth factors and matricellular proteins and interactions with other stromal cells. While weight loss is recommended to resolve obesity, the long-term effects of weight loss on adipose tissue fibrosis and inflammation within breast tissue are less clear. Increased fibrosis within breast tissue may increase the risk for tumor development as well as promote characteristics associated with tumor aggressiveness.
Collapse
Affiliation(s)
- Genevra Kuziel
- Cancer Biology Graduate Program, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
| | - Brittney N Moore
- Department of Comparative Biosciences, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706, USA
| | - Lisa M Arendt
- Cancer Biology Graduate Program, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
- Department of Comparative Biosciences, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706, USA
| |
Collapse
|
21
|
Zhou Y, Ye Z, Wei W, Zhang M, Huang F, Li J, Cai C. Macrophages maintain mammary stem cell activity and mammary homeostasis via TNF-α-PI3K-Cdk1/Cyclin B1 axis. NPJ Regen Med 2023; 8:23. [PMID: 37130846 PMCID: PMC10154328 DOI: 10.1038/s41536-023-00296-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 04/20/2023] [Indexed: 05/04/2023] Open
Abstract
Adult stem cell niche is a special environment composed of a variety stromal cells and signals, which cooperatively regulate tissue development and homeostasis. It is of great interest to study the role of immune cells in niche. Here, we show that mammary resident macrophages regulate mammary epithelium cell division and mammary development through TNF-α-Cdk1/Cyclin B1 axis. In vivo, depletion of macrophages reduces the number of mammary basal cells and mammary stem cells (MaSCs), while increases mammary luminal cells. In vitro, we establish a three-dimensional culture system in which mammary basal cells are co-cultured with macrophages, and interestingly, macrophage co-culture promotes the formation of branched functional mammary organoids. Moreover, TNF-α produced by macrophages activates the intracellular PI3K/Cdk1/Cyclin B1 signaling in mammary cells, thereby maintaining the activity of MaSCs and the formation of mammary organoids. Together, these findings reveal the functional significance of macrophageal niche and intracellular PI3K/Cdk1/Cyclin B1 axis for maintaining MaSC activity and mammary homeostasis.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zi Ye
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Wei Wei
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Mengna Zhang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Fujing Huang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jinpeng Li
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China.
| | - Cheguo Cai
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China.
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| |
Collapse
|
22
|
Hirano R, Okamoto K, Shinke M, Sato M, Watanabe S, Watanabe H, Kondoh G, Kadonosono T, Kizaka-Kondoh S. Tissue-resident macrophages are major tumor-associated macrophage resources, contributing to early TNBC development, recurrence, and metastases. Commun Biol 2023; 6:144. [PMID: 36737474 PMCID: PMC9898263 DOI: 10.1038/s42003-023-04525-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and highly heterogenous disease with no well-defined therapeutic targets. Treatment options are thus limited and mortality is significantly higher compared with other breast cancer subtypes. Mammary gland tissue-resident macrophages (MGTRMs) are found to be the most abundant stromal cells in early TNBC before angiogenesis. We therefore aimed to explore novel therapeutic approaches for TNBC by focusing on MGTRMs. Local depletion of MGTRMs in mammary gland fat pads the day before TNBC cell transplantation significantly reduced tumor growth and tumor-associated macrophage (TAM) infiltration in mice. Furthermore, local depletion of MGTRMs at the site of TNBC resection markedly reduced recurrence and distant metastases, and improved chemotherapy outcomes. This study demonstrates that MGTRMs are a major TAM resource and play pivotal roles in the growth and malignant progression of TNBC. The results highlight a possible novel anti-cancer approach targeting tissue-resident macrophages.
Collapse
Affiliation(s)
- Ryuichiro Hirano
- grid.32197.3e0000 0001 2179 2105School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Koki Okamoto
- grid.32197.3e0000 0001 2179 2105School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Miyu Shinke
- grid.32197.3e0000 0001 2179 2105School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Marika Sato
- grid.32197.3e0000 0001 2179 2105School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Shigeaki Watanabe
- grid.32197.3e0000 0001 2179 2105School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Hitomi Watanabe
- grid.258799.80000 0004 0372 2033Institute for Life and Medical Sciences, Kyoto University, Sakyo, Kyoto, 606-8507 Japan
| | - Gen Kondoh
- grid.258799.80000 0004 0372 2033Institute for Life and Medical Sciences, Kyoto University, Sakyo, Kyoto, 606-8507 Japan
| | - Tetsuya Kadonosono
- grid.32197.3e0000 0001 2179 2105School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Shinae Kizaka-Kondoh
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan.
| |
Collapse
|
23
|
Zhang X, Ji L, Li MO. Control of tumor-associated macrophage responses by nutrient acquisition and metabolism. Immunity 2023; 56:14-31. [PMID: 36630912 PMCID: PMC9839308 DOI: 10.1016/j.immuni.2022.12.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/29/2022] [Accepted: 12/06/2022] [Indexed: 01/11/2023]
Abstract
Metazoan tissue specification is associated with integration of macrophage lineage cells in sub-tissular niches to promote tissue development and homeostasis. Oncogenic transformation, most prevalently of epithelial cell lineages, results in maladaptation of resident tissue macrophage differentiation pathways to generate parenchymal and interstitial tumor-associated macrophages that largely foster cancer progression. In addition to growth factors, nutrients that can be consumed, stored, recycled, or converted to signaling molecules have emerged as crucial regulators of macrophage responses in tumor. Here, we review how nutrient acquisition through plasma membrane transporters and engulfment pathways control tumor-associated macrophage differentiation and function. We also discuss how nutrient metabolism regulates tumor-associated macrophages and how these processes may be targeted for cancer therapy.
Collapse
Affiliation(s)
- Xian Zhang
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Liangliang Ji
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ming O Li
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA.
| |
Collapse
|
24
|
Li N, Li Z, Fang F, Zhu C, Zhang W, Lu Y, Zhang R, Si P, Bian Y, Qin Y, Jiao X. Two distinct resident macrophage populations coexist in the ovary. Front Immunol 2022; 13:1007711. [PMID: 36605192 PMCID: PMC9810109 DOI: 10.3389/fimmu.2022.1007711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction Tissue-resident macrophages (TRMs) are highly heterogeneous and have a complex and important role in tissue support, homeostasis, and function. The heterogeneity, maintenance, and function of TRMs, as one of the major immune cells in the ovary, are not well understood. Methods Application of flow cytometry, Parabiosis, Fate mapping, Macrophage depletion, etc. Results Here, we described two distinct macrophage subsets, F4/80hiCD11bint and F4/80intCD11bhi, with different phenotypic characteristics in the ovary of mice. The F4/80hiCD11bint population contained a distinct CD206+ subgroup and highly expressed CD81, while the F4/80intCD11bhi subset showed higher expression of CCR2 and TLR2. Notably, Ly6c+ macrophages were present almost exclusively in the F4/80intCD11bhi subpopulation. Combining in vivo fate mapping and parabiotic mouse models, we characterized the longevity and replenishment of the two macrophage populations. We found that both the F4/80hiCD11bint and F4/80intCD11bhi subsets were ovary-resident. Importantly, the F4/80hiCD11bint macrophages acted as a self-maintaining and long-lived population with a modest monocyte contribution at a steady state, and the F4/80intCD11bhi subpopulation had a relatively short lifespan with a greater contribution from monocytes. After macrophage ablation, disturbance of estradiol secretion and ovarian hemorrhage due to damaged vascular integrity was observed in mice. Discussion Our data provide critical insights into ovarian macrophage heterogeneity and highlight the strategic role of TRMs in ovarian homeostasis and physiology.
Collapse
Affiliation(s)
- Nianyu Li
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China,Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, China,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China,Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Zhuqing Li
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China,Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, China,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China,Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Fang Fang
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China,Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, China,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China,Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Chendi Zhu
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China,Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, China,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China,Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Wenzhe Zhang
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China,Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, China,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China,Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Yueshuang Lu
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China,Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, China,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China,Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Rongrong Zhang
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China,Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, China,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China,Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Pinxin Si
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China,Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, China,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China,Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Yuehong Bian
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China,Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, China,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China,Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Yingying Qin
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China,Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, China,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China,Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Xue Jiao
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China,Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, China,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China,Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China,Suzhou Institute of Shandong University, Suzhou, Jiangsu, China,*Correspondence: Xue Jiao,
| |
Collapse
|
25
|
Alkhattabi NA, Hussein SA, Tarbiah NI, Alzahri RY, Khalifa R. Thymoquinone Effect on Monocyte-Derived Macrophages, Cell-Surface Molecule Expression, and Phagocytosis. Nutrients 2022; 14:nu14245240. [PMID: 36558399 PMCID: PMC9783248 DOI: 10.3390/nu14245240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 11/27/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Macrophages are one of the most important cells in the immune system. They act as links between innate and adaptive immunities. In this study, the aim was to examine thymoquinone effects on the immunological properties of different macrophages. Peripheral blood mononuclear cells were isolated from blood from healthy volunteers by negative selection of monocytes that had been cultured for seven days to differentiate into macrophages. Cells were cultured with or without the presence of thymoquinone (TQ), which was used in two different concentrations (50 μg/mL and 100 μg/mL. Cluster of differentiation 80 (CD80), cluster of differentiation 86 (CD86), and human leukocyte antigen DR isotype (HLA-DR) were measured by flow cytometry, and the secretion of interferon gamma (IFN-γ) and tumour necrosis factor alpha (TNF-α) was measured. Cells were also tested for their E. coli phagocytosis abilities. The data showed that the expression of HLA-DR was significantly higher in cells treated with 100 μL/mL TQ. In addition, IFN-γ concentration increased in the 100 μg/mL TQ-treated cells. The macrophage phagocytosis results showed a significant difference in 50 μg/mL TQ-treated cells compared to the controls. TQ may enhance the immunological properties of macrophages during the early stages of innate immunity by activating phagocytosis ability and by increasing the expression of HLA-DR and the secretion of IFN-γ, which may enhance the antigen-presentation capabilities of macrophages.
Collapse
Affiliation(s)
- Nuha A. Alkhattabi
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Correspondence: ; Tel.: +966-536665958
| | - Sowsan A. Hussein
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Nesrin I. Tarbiah
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Reem Y. Alzahri
- Department of Biology, College of Science, University of Jeddah, Jeddah 21493, Saudi Arabia
| | - Reham Khalifa
- Medical Microbiology and Immunology, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| |
Collapse
|
26
|
The origins of resident macrophages in mammary gland influence the tumorigenesis of breast cancer. Int Immunopharmacol 2022; 110:109047. [DOI: 10.1016/j.intimp.2022.109047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 11/05/2022]
|
27
|
Laviron M, Petit M, Weber-Delacroix E, Combes AJ, Arkal AR, Barthélémy S, Courau T, Hume DA, Combadière C, Krummel MF, Boissonnas A. Tumor-associated macrophage heterogeneity is driven by tissue territories in breast cancer. Cell Rep 2022; 39:110865. [PMID: 35613577 DOI: 10.1016/j.celrep.2022.110865] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 04/04/2022] [Accepted: 05/03/2022] [Indexed: 02/08/2023] Open
Abstract
Tissue-resident macrophages adapt to local signals within tissues to acquire specific functions. Neoplasia transforms the tissue, raising the question as to how the environmental perturbations contribute to tumor-associated macrophage (TAM) identity and functions. Combining single-cell RNA sequencing (scRNA-seq) with spatial localization of distinct TAM subsets by imaging, we discover that TAM transcriptomic programs follow two main differentiation paths according to their localization in the stroma or in the neoplastic epithelium of the mammary duct. Furthermore, this diversity is exclusively detected in a spontaneous tumor model and tracks the different tissue territories as well as the type of tumor lesion. These TAM subsets harbor distinct capacity to activate CD8+ T cells and phagocyte tumor cells, supporting that specific tumor regions, rather than defined activation states, are the major drivers of TAM plasticity and heterogeneity. The distinctions created here provide a framework to design cancer treatment targeting specific TAM niches.
Collapse
Affiliation(s)
- Marie Laviron
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, 75013 Paris, France
| | - Maxime Petit
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, 75013 Paris, France
| | - Eléonore Weber-Delacroix
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, 75013 Paris, France
| | - Alexis J Combes
- Department of Pathology, ImmunoX Initiative, UCSF Immunoprofiler Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arjun Rao Arkal
- Department of Pathology, ImmunoX Initiative, UCSF Immunoprofiler Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sandrine Barthélémy
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, 75013 Paris, France
| | - Tristan Courau
- Department of Pathology, ImmunoX Initiative, UCSF Immunoprofiler Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David A Hume
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, QLD 4101, Australia
| | - Christophe Combadière
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, 75013 Paris, France
| | - Matthew F Krummel
- Department of Pathology, ImmunoX Initiative, UCSF Immunoprofiler Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alexandre Boissonnas
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, 75013 Paris, France.
| |
Collapse
|
28
|
Hitchcock J, Hughes K, Pensa S, Lloyd-Lewis B, Watson CJ. The immune environment of the mammary gland fluctuates during post-lactational regression and correlates with tumour growth rate. Development 2022; 149:275060. [PMID: 35420674 PMCID: PMC9124574 DOI: 10.1242/dev.200162] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 04/04/2022] [Indexed: 01/02/2023]
Abstract
Post-lactational mammary gland regression encompasses extensive programmed cell death and removal of milk-producing epithelial cells, breakdown of extracellular matrix components and redifferentiation of stromal adipocytes. This highly regulated involution process is associated with a transient increased risk of breast cancer in women. Using a syngeneic tumour model, we show that tumour growth is significantly altered depending on the stage of involution at which tumour cells are implanted. Tumour cells injected at day 3 involution grew faster than those in nulliparous mice, whereas tumours initiated at day 6 involution grew significantly slower. These differences in tumour progression correlate with distinct changes in innate immune cells, in particular among F4/80-expressing macrophages and among TCRδ+ unconventional T cells. Breast cancer post-pregnancy risk is exacerbated in older first-time mothers and, in our model, initial tumour growth is moderately faster in aged mice compared with young mice. Our results have implications for breast cancer risk and the use of anti-inflammatory therapeutics for postpartum breast cancers. Summary: Mammary gland involution is associated with dynamic changes in immune cell types and numbers at different stages that correlates with the initial rate of growth of implanted tumour cells.
Collapse
Affiliation(s)
- Jessica Hitchcock
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Katherine Hughes
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK
| | - Sara Pensa
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Bethan Lloyd-Lewis
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
- School of Cellular and Molecular Medicine, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - Christine J. Watson
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| |
Collapse
|
29
|
Abstract
Although best known for their phagocytic and immunological functions, macrophages have increasingly been recognised as key players in the development, homeostasis and regeneration of their host tissues. Early during development, macrophages infiltrate and colonise all tissues within the body, developing symbiotically with their host tissues and acquiring unique functional adaptations based on the tissue microenvironment. These embryonic resident tissue macrophages (RTMs) are ontogenically distinct from the later adult bone marrow-derived monocytes, and in some tissues are self-maintained independently of general circulation at a steady state. In this article, we briefly discuss the ontogeny, maintenance and unique tissue adaptions of RTMs focusing on microglia, Kupffer cells, Langerhans cells, intestinal macrophages, cardiac macrophages and tumour-associated macrophages, and highlight their role in development, homeostasis and dysfunction.
Collapse
Affiliation(s)
- Christopher Zhe Wei Lee
- Singapore Immunology Network, 8A Biomedical Grove, Singapore 138648, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Florent Ginhoux
- Singapore Immunology Network, 8A Biomedical Grove, Singapore 138648, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore.,Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, MD9-04-07, 2 Medical Drive, Singapore 117593, Singapore.,Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, 31 Third Hospital Ave, #03-03 Bowyer Block C, Singapore 168753, Singapore
| |
Collapse
|
30
|
Nalio Ramos R, Missolo-Koussou Y, Gerber-Ferder Y, Bromley CP, Bugatti M, Núñez NG, Tosello Boari J, Richer W, Menger L, Denizeau J, Sedlik C, Caudana P, Kotsias F, Niborski LL, Viel S, Bohec M, Lameiras S, Baulande S, Lesage L, Nicolas A, Meseure D, Vincent-Salomon A, Reyal F, Dutertre CA, Ginhoux F, Vimeux L, Donnadieu E, Buttard B, Galon J, Zelenay S, Vermi W, Guermonprez P, Piaggio E, Helft J. Tissue-resident FOLR2 + macrophages associate with CD8 + T cell infiltration in human breast cancer. Cell 2022; 185:1189-1207.e25. [PMID: 35325594 DOI: 10.1016/j.cell.2022.02.021] [Citation(s) in RCA: 271] [Impact Index Per Article: 90.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 10/08/2021] [Accepted: 02/15/2022] [Indexed: 12/12/2022]
Abstract
Macrophage infiltration is a hallmark of solid cancers, and overall macrophage infiltration correlates with lower patient survival and resistance to therapy. Tumor-associated macrophages, however, are phenotypically and functionally heterogeneous. Specific subsets of tumor-associated macrophage might be endowed with distinct roles on cancer progression and antitumor immunity. Here, we identify a discrete population of FOLR2+ tissue-resident macrophages in healthy mammary gland and breast cancer primary tumors. FOLR2+ macrophages localize in perivascular areas in the tumor stroma, where they interact with CD8+ T cells. FOLR2+ macrophages efficiently prime effector CD8+ T cells ex vivo. The density of FOLR2+ macrophages in tumors positively correlates with better patient survival. This study highlights specific roles for tumor-associated macrophage subsets and paves the way for subset-targeted therapeutic interventions in macrophages-based cancer therapies.
Collapse
Affiliation(s)
- Rodrigo Nalio Ramos
- PSL University, Institut Curie Research Center, INSERM U932 & SiRIC, Translational Immunotherapy Team, 75005 Paris, France
| | - Yoann Missolo-Koussou
- PSL University, Institut Curie Research Center, INSERM U932 & SiRIC, Translational Immunotherapy Team, 75005 Paris, France
| | - Yohan Gerber-Ferder
- PSL University, Institut Curie Research Center, INSERM U932 & SiRIC, Translational Immunotherapy Team, 75005 Paris, France
| | - Christian P Bromley
- Cancer Inflammation and Immunity Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Manchester, UK
| | - Mattia Bugatti
- Department of Pathology, University of Brescia, Brescia 25123, Italy
| | - Nicolas Gonzalo Núñez
- PSL University, Institut Curie Research Center, INSERM U932 & SiRIC, Translational Immunotherapy Team, 75005 Paris, France
| | - Jimena Tosello Boari
- PSL University, Institut Curie Research Center, INSERM U932 & SiRIC, Translational Immunotherapy Team, 75005 Paris, France
| | - Wilfrid Richer
- PSL University, Institut Curie Research Center, INSERM U932 & SiRIC, Translational Immunotherapy Team, 75005 Paris, France
| | - Laurie Menger
- PSL University, Institut Curie Research Center, INSERM U932, 75005 Paris, France
| | - Jordan Denizeau
- PSL University, Institut Curie Research Center, INSERM U932 & SiRIC, Translational Immunotherapy Team, 75005 Paris, France
| | - Christine Sedlik
- PSL University, Institut Curie Research Center, INSERM U932 & SiRIC, Translational Immunotherapy Team, 75005 Paris, France
| | - Pamela Caudana
- PSL University, Institut Curie Research Center, INSERM U932 & SiRIC, Translational Immunotherapy Team, 75005 Paris, France
| | - Fiorella Kotsias
- PSL University, Institut Curie Research Center, INSERM U932 & SiRIC, Translational Immunotherapy Team, 75005 Paris, France
| | - Leticia L Niborski
- PSL University, Institut Curie Research Center, INSERM U932 & SiRIC, Translational Immunotherapy Team, 75005 Paris, France
| | - Sophie Viel
- PSL University, Institut Curie Research Center, INSERM U932 & SiRIC, Translational Immunotherapy Team, 75005 Paris, France
| | - Mylène Bohec
- PSL University, Institut Curie Research Center, Institut Curie Genomics of Excellence Platform, 75005 Paris, France
| | - Sonia Lameiras
- PSL University, Institut Curie Research Center, Institut Curie Genomics of Excellence Platform, 75005 Paris, France
| | - Sylvain Baulande
- PSL University, Institut Curie Research Center, Institut Curie Genomics of Excellence Platform, 75005 Paris, France
| | - Laëtitia Lesage
- PSL University, Institut Curie Hospital, Department of Pathology, 75005 Paris, France
| | - André Nicolas
- PSL University, Institut Curie Hospital, Department of Pathology, 75005 Paris, France
| | - Didier Meseure
- PSL University, Institut Curie Hospital, Department of Pathology, 75005 Paris, France
| | - Anne Vincent-Salomon
- PSL University, Institut Curie Hospital, Department of Pathology, 75005 Paris, France
| | - Fabien Reyal
- PSL University, Institut Curie Hospital, Department of Surgery, 75005 Paris, France
| | | | - Florent Ginhoux
- Université Paris-Saclay, Institut Gustave Roussy, INSERM U1015, Villejuif, France; Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648, Singapore
| | - Lene Vimeux
- University of Paris, Institut Cochin, INSERM U1016, CNRS UMR 8104, 75014 Paris, France
| | - Emmanuel Donnadieu
- University of Paris, Institut Cochin, INSERM U1016, CNRS UMR 8104, 75014 Paris, France
| | - Bénédicte Buttard
- INSERM, Sorbonne Université, Université de Paris, Centre de Recherche des Cordeliers, Laboratory of Integrative Cancer Immunology, Paris, France
| | - Jérôme Galon
- INSERM, Sorbonne Université, Université de Paris, Centre de Recherche des Cordeliers, Laboratory of Integrative Cancer Immunology, Paris, France
| | - Santiago Zelenay
- Cancer Inflammation and Immunity Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Manchester, UK
| | - William Vermi
- PSL University, Institut Curie Research Center, INSERM U932, 75005 Paris, France; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Pierre Guermonprez
- Université de Paris, Centre for Inflammation Research, CNRS ERL8252, INSERM1149, Paris, France
| | - Eliane Piaggio
- PSL University, Institut Curie Research Center, INSERM U932 & SiRIC, Translational Immunotherapy Team, 75005 Paris, France
| | - Julie Helft
- PSL University, Institut Curie Research Center, INSERM U932 & SiRIC, Translational Immunotherapy Team, 75005 Paris, France.
| |
Collapse
|
31
|
Hadadi E, Deschoemaeker S, Vicente Venegas G, Laoui D. Heterogeneity and function of macrophages in the breast during homeostasis and cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 367:149-182. [PMID: 35461657 DOI: 10.1016/bs.ircmb.2022.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Macrophages are diverse immune cells populating all tissues and adopting a unique tissue-specific identity. Breast macrophages play an essential role in the development and function of the mammary gland over one's lifetime. In the recent years, with the development of fate-mapping, imaging and scRNA-seq technologies we grew a better understanding of the origin, heterogeneity and function of mammary macrophages in homeostasis but also during breast cancer development. Here, we aim to provide a comprehensive review of the latest improvements in studying the macrophage heterogeneity in healthy mammary tissues and breast cancer.
Collapse
Affiliation(s)
- Eva Hadadi
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sofie Deschoemaeker
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Gerard Vicente Venegas
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Damya Laoui
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
32
|
Differential expression of CD11c defines two types of tissue-resident macrophages with different origins in steady-state salivary glands. Sci Rep 2022; 12:931. [PMID: 35042931 PMCID: PMC8766464 DOI: 10.1038/s41598-022-04941-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 01/04/2022] [Indexed: 12/24/2022] Open
Abstract
Gland macrophages are primed for gland development and functions through interactions within their niche. However, the phenotype, ontogeny, and function of steady-state salivary gland (SG) macrophages remain unclear. We herein identified CD11c+ and CD11c- subsets among CD64+ macrophages in steady-state murine SGs. CD11c- macrophages were predominant in the SGs of embryonic and newborn mice and decreased with advancing age. CD11c+ macrophages were rarely detected in the embryonic period, but rapidly expanded after birth. CD11c+, but not CD11c-, macrophage numbers decreased in mice treated with a CCR2 antagonist, suggesting that CD11c+ macrophages accumulate from bone marrow-derived progenitors in a CCR2-dependent manner, whereas CD11c- macrophages were derived from embryonic progenitors in SGs. CD11c+ and CD11c- macrophages strongly expressed colony-stimulating factor (CSF)-1 receptor, the injection of an anti-CSF-1 receptor blocking antibody markedly reduced both subsets, and SGs strongly expressed CSF-1, indicating the dependency of SG resident macrophage development on CSF-1. The phagocytic activity of SG macrophages was extremely weak; however, the gene expression profile of SG macrophages indicated that SG macrophages regulate gland development and functions in SGs. These results suggest that SG CD11c+ and CD11c- macrophages are developed and instructed to perform SG-specific functions in steady-state SGs.
Collapse
|
33
|
McKendrick JG, Emmerson E. The role of salivary gland macrophages in infection, disease and repair. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 368:1-34. [PMID: 35636925 DOI: 10.1016/bs.ircmb.2022.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Macrophages are mononuclear innate immune cells which have become of increasing interest in the fields of disease and regeneration, as their non-classical functions have been elucidated in addition to their classical inflammatory functions. Macrophages can regulate tissue remodeling, by both mounting and reducing inflammatory responses; and exhibit direct communication with other cells to drive tissue turnover and cell replacement. Furthermore, macrophages have recently become an attractive therapeutic target to drive tissue regeneration. The major salivary glands are glandular tissues that are exposed to pathogens through their close connection with the oral cavity. Moreover, there are a number of diseases that preferentially destroy the salivary glands, causing irreversible injury, highlighting the need for a regenerative strategy. However, characterization of macrophages in the mouse and human salivary glands is sparse and has been mostly determined from studies in infection or autoimmune pathologies. In this review, we describe the current literature around salivary gland macrophages, and speculate about the niches they inhabit and how their role in development, regeneration and cancer may inform future therapeutic advances.
Collapse
Affiliation(s)
- John G McKendrick
- The Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom
| | - Elaine Emmerson
- The Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
34
|
Wilson GJ, Fukuoka A, Vidler F, Graham GJ. Diverse myeloid cells are recruited to the developing and inflamed mammary gland. Immunology 2021; 165:206-218. [PMID: 34775606 DOI: 10.1111/imm.13430] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 10/27/2021] [Accepted: 11/09/2021] [Indexed: 12/26/2022] Open
Abstract
The immune system plays fundamental roles in the mammary gland, shaping developmental processes and controlling inflammation during infection and cancer. Here, we reveal unanticipated heterogeneity in the myeloid cell compartment during development of virgin, pregnant, lactating and involuting mouse mammary glands, and in milk. We investigate the functional consequences of individual and compound chemokine receptor deficiency on cell recruitment. Diverse myeloid cell recruitment was also shown in models of sterile inflammation and bacterial infection. Strikingly, we have shown that inflammation and infection can alter the abundance of terminal end buds, a key developmental structure, within the pubertal mammary gland. This previously unknown effect of inflammatory burden during puberty could have important implications for understanding pubertal development.
Collapse
Affiliation(s)
- Gillian J Wilson
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Ayumi Fukuoka
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Francesca Vidler
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Gerard J Graham
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| |
Collapse
|
35
|
Nagy D, Gillis CMC, Davies K, Fowden AL, Rees P, Wills JW, Hughes K. Developing ovine mammary terminal duct lobular units have a dynamic mucosal and stromal immune microenvironment. Commun Biol 2021; 4:993. [PMID: 34417554 PMCID: PMC8379191 DOI: 10.1038/s42003-021-02502-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 07/27/2021] [Indexed: 11/29/2022] Open
Abstract
The human breast and ovine mammary gland undergo striking levels of postnatal development, leading to formation of terminal duct lobular units (TDLUs). Here we interrogate aspects of sheep TDLU growth as a model of breast development and to increase understanding of ovine mammogenesis. The distributions of epithelial nuclear Ki67 positivity differ significantly between younger and older lambs. Ki67 expression is polarised to the leading edge of the developing TDLUs. Intraepithelial ductal macrophages exhibit periodicity and considerably increased density in lambs approaching puberty. Stromal macrophages are more abundant centrally than peripherally. Intraepithelial T lymphocytes are more numerous in older lambs. Stromal hotspots of Ki67 expression colocalize with immune cell aggregates that exhibit distinct organisation consistent with tertiary lymphoid structures. The lamb mammary gland thus exhibits a dynamic mucosal and stromal immune microenvironment and constitutes a valuable model system that provides new insights into postnatal breast development.
Collapse
Affiliation(s)
- Dorottya Nagy
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
- Equine Clinic, Department of Companion Animals and Equids, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Clare M C Gillis
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Katie Davies
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK
| | - Abigail L Fowden
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK
| | - Paul Rees
- College of Engineering, Swansea University, Fabian Way, Crymlyn Burrows, Swansea, UK
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - John W Wills
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK.
| | - Katherine Hughes
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
36
|
Félix I, Jokela H, Karhula J, Kotaja N, Savontaus E, Salmi M, Rantakari P. Single-Cell Proteomics Reveals the Defined Heterogeneity of Resident Macrophages in White Adipose Tissue. Front Immunol 2021; 12:719979. [PMID: 34381461 PMCID: PMC8350344 DOI: 10.3389/fimmu.2021.719979] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 07/12/2021] [Indexed: 12/24/2022] Open
Abstract
Adipose tissue macrophages (ATMs) regulate homeostasis and contribute to the metabolically harmful chronic inflammation in obese individuals. While evident heterogeneity of resident ATMs has been described previously, their phenotype, developmental origin, and functionality remain inconsistent. We analyzed white adipose tissue (WAT) during homeostasis and diet interventions using comprehensive and unbiased single-cell mass cytometry and genetic lineage tracking models. We now provide a uniform definition of individual subsets of resident ATMs. We show that in lean mice, WAT co-harbors eight kinetically evolving CD206+ macrophage subpopulations (defined by TIM4, CD163, and MHC II) and two CD206- macrophage subpopulations. TIM4-CD163+, TIM4-CD163- and CD206- macrophage populations are largely bone marrow-derived, while the proliferating TIM4+CD163+ subpopulation is of embryonic origin. All macrophage subtypes are active in phagocytosis, endocytosis, and antigen processing in vitro, whereas TIM4+CD163+ cells are superior in scavenging in vivo. A high-fat diet induces massive infiltration of CD206- macrophages and selective down-regulation of MHC II on TIM4+ macrophages. These changes are reversed by dietary intervention. Thus, the developmental origin and environment jointly regulate the functional malleability of resident ATMs.
Collapse
Affiliation(s)
- Inês Félix
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.,Research Centre for Infection and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland.,InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Heli Jokela
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.,Research Centre for Infection and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland.,InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Joonas Karhula
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.,Research Centre for Infection and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland.,InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Noora Kotaja
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Eriika Savontaus
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.,Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Marko Salmi
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Pia Rantakari
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.,Research Centre for Infection and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland.,InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| |
Collapse
|
37
|
Li A, Zhao Y, Li Y, Jiang L, Gu Y, Liu J. Cell-derived biomimetic nanocarriers for targeted cancer therapy: cell membranes and extracellular vesicles. Drug Deliv 2021; 28:1237-1255. [PMID: 34142930 PMCID: PMC8216268 DOI: 10.1080/10717544.2021.1938757] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nanotechnology provides synthetic carriers for cancer drug delivery that protect cargos from degradation, control drug release and increase local accumulation at tumors. However, these non-natural vehicles display poor tumor targeting and potential toxicity and are eliminated by the immune system. Recently, biomimetic nanocarriers have been widely developed based on the concept of ‘mimicking nature.’ Among them, cell-derived biomimetic vehicles have become the focus of bionics research because of their multiple natural functions, such as low immunogenicity, long circulation time and targeting ability. Cell membrane-coated carriers and extracellular vesicles are two widely used cell-based biomimetic materials. Here, this review summarizes the latest progress in the application of these two biomimetic carriers in targeted cancer therapy. Their properties and performance are compared, and their future challenges and development prospects are discussed.
Collapse
Affiliation(s)
- Aixue Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.,Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yunan Zhao
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yixiu Li
- Department of Pharmacy, Shanghai Integrated Traditional Chinese and Western Medicine Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liangdi Jiang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.,Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yongwei Gu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiyong Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
38
|
Slepicka PF, Somasundara AVH, Dos Santos CO. The molecular basis of mammary gland development and epithelial differentiation. Semin Cell Dev Biol 2021; 114:93-112. [PMID: 33082117 PMCID: PMC8052380 DOI: 10.1016/j.semcdb.2020.09.014] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/28/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023]
Abstract
Our understanding of the molecular events underpinning the development of mammalian organ systems has been increasing rapidly in recent years. With the advent of new and improved next-generation sequencing methods, we are now able to dig deeper than ever before into the genomic and epigenomic events that play critical roles in determining the fates of stem and progenitor cells during the development of an embryo into an adult. In this review, we detail and discuss the genes and pathways that are involved in mammary gland development, from embryogenesis, through maturation into an adult gland, to the role of pregnancy signals in directing the terminal maturation of the mammary gland into a milk producing organ that can nurture the offspring. We also provide an overview of the latest research in the single-cell genomics of mammary gland development, which may help us to understand the lineage commitment of mammary stem cells (MaSCs) into luminal or basal epithelial cells that constitute the mammary gland. Finally, we summarize the use of 3D organoid cultures as a model system to study the molecular events during mammary gland development. Our increased investigation of the molecular requirements for normal mammary gland development will advance the discovery of targets to predict breast cancer risk and the development of new breast cancer therapies.
Collapse
Affiliation(s)
- Priscila Ferreira Slepicka
- Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | | | - Camila O Dos Santos
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
39
|
Cotechini T, Atallah A, Grossman A. Tissue-Resident and Recruited Macrophages in Primary Tumor and Metastatic Microenvironments: Potential Targets in Cancer Therapy. Cells 2021; 10:960. [PMID: 33924237 PMCID: PMC8074766 DOI: 10.3390/cells10040960] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 12/24/2022] Open
Abstract
Macrophages within solid tumors and metastatic sites are heterogenous populations with different developmental origins and substantially contribute to tumor progression. A number of tumor-promoting phenotypes associated with both tumor- and metastasis-associated macrophages are similar to innate programs of embryonic-derived tissue-resident macrophages. In contrast to recruited macrophages originating from marrow precursors, tissue-resident macrophages are seeded before birth and function to coordinate tissue remodeling and maintain tissue integrity and homeostasis. Both recruited and tissue-resident macrophage populations contribute to tumor growth and metastasis and are important mediators of resistance to chemotherapy, radiation therapy, and immune checkpoint blockade. Thus, targeting various macrophage populations and their tumor-promoting phenotypes holds therapeutic promise. Here, we discuss various macrophage populations as regulators of tumor progression, immunity, and immunotherapy. We provide an overview of macrophage targeting strategies, including therapeutics designed to induce macrophage depletion, impair recruitment, and induce repolarization. We also provide a perspective on the therapeutic potential for macrophage-specific acquisition of trained immunity as an anti-cancer agent and discuss the therapeutic potential of exploiting macrophages and their traits to reduce tumor burden.
Collapse
Affiliation(s)
- Tiziana Cotechini
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (A.A.); (A.G.)
| | | | | |
Collapse
|
40
|
Zhang XM, Chen DG, Li SC, Zhu B, Li ZJ. Embryonic Origin and Subclonal Evolution of Tumor-Associated Macrophages Imply Preventive Care for Cancer. Cells 2021; 10:903. [PMID: 33919979 PMCID: PMC8071014 DOI: 10.3390/cells10040903] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 01/16/2023] Open
Abstract
Macrophages are widely distributed in tissues and function in homeostasis. During cancer development, tumor-associated macrophages (TAMs) dominatingly support disease progression and resistance to therapy by promoting tumor proliferation, angiogenesis, metastasis, and immunosuppression, thereby making TAMs a target for tumor immunotherapy. Here, we started with evidence that TAMs are highly plastic and heterogeneous in phenotype and function in response to microenvironmental cues. We pointed out that efforts to tear off the heterogeneous "camouflage" in TAMs conduce to target de facto protumoral TAMs efficiently. In particular, several fate-mapping models suggest that most tissue-resident macrophages (TRMs) are generated from embryonic progenitors, and new paradigms uncover the ontogeny of TAMs. First, TAMs from embryonic modeling of TRMs and circulating monocytes have distinct transcriptional profiling and function, suggesting that the ontogeny of TAMs is responsible for the functional heterogeneity of TAMs, in addition to microenvironmental cues. Second, metabolic remodeling helps determine the mechanism of phenotypic and functional characteristics in TAMs, including metabolic bias from macrophages' ontogeny in macrophages' functional plasticity under physiological and pathological conditions. Both models aim at dissecting the ontogeny-related metabolic regulation in the phenotypic and functional heterogeneity in TAMs. We argue that gleaning from the single-cell transcriptomics on subclonal TAMs' origins may help understand the classification of TAMs' population in subclonal evolution and their distinct roles in tumor development. We envision that TAM-subclone-specific metabolic reprogramming may round-up with future cancer therapies.
Collapse
Affiliation(s)
- Xiao-Mei Zhang
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing 400037, China;
| | - De-Gao Chen
- Institute of Cancer, The Second Affiliated Hospital, Army Military Medical University, Chongqing 400037, China;
| | - Shengwen Calvin Li
- Neuro-Oncology and Stem Cell Research Laboratory, Center for Neuroscience Research, CHOC Children’s Research Institute, Children’s Hospital of Orange County (CHOC), 1201 West La Veta Ave., Orange, CA 92868, USA
- Department of Neurology, University of California-Irvine School of Medicine, 200 S Manchester Ave., Ste 206, Orange, CA 92868, USA
| | - Bo Zhu
- Institute of Cancer, The Second Affiliated Hospital, Army Military Medical University, Chongqing 400037, China;
| | - Zhong-Jun Li
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing 400037, China;
| |
Collapse
|
41
|
Gu X, Li SY, DeFalco T. Immune and vascular contributions to organogenesis of the testis and ovary. FEBS J 2021; 289:2386-2408. [PMID: 33774913 PMCID: PMC8476657 DOI: 10.1111/febs.15848] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/07/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023]
Abstract
Gonad development is a highly regulated process that coordinates cell specification and morphogenesis to produce sex-specific organ structures that are required for fertility, such as testicular seminiferous tubules and ovarian follicles. While sex determination occurs within specialized gonadal supporting cells, sexual differentiation is evident throughout the entire organ, including within the interstitial compartment, which contains immune cells and vasculature. While immune and vascular cells have been traditionally appreciated for their supporting roles during tissue growth and homeostasis, an increasing body of evidence supports the idea that these cell types are critical drivers of sexually dimorphic morphogenesis of the gonad. Myeloid immune cells, such as macrophages, are essential for multiple aspects of gonadogenesis and fertility, including for forming and maintaining gonadal vasculature in both sexes at varying stages of life. While vasculature is long known for supporting organ growth and serving as an export mechanism for gonadal sex steroids in utero, it is also an important component of fetal testicular morphogenesis and differentiation; additionally, it is vital for ovarian corpus luteal function and maintenance of pregnancy. These findings point toward a new paradigm in which immune cells and blood vessels are integral components of sexual differentiation and organogenesis. In this review, we discuss the state of the field regarding the diverse roles of immune and vascular cells during organogenesis of the testis and ovary and highlight outstanding questions in the field that could stimulate new research into these previously underappreciated constituents of the gonad.
Collapse
Affiliation(s)
- Xiaowei Gu
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Shu-Yun Li
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Tony DeFalco
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, OH, USA
| |
Collapse
|
42
|
Bach K, Pensa S, Zarocsinceva M, Kania K, Stockis J, Pinaud S, Lazarus KA, Shehata M, Simões BM, Greenhalgh AR, Howell SJ, Clarke RB, Caldas C, Halim TYF, Marioni JC, Khaled WT. Time-resolved single-cell analysis of Brca1 associated mammary tumourigenesis reveals aberrant differentiation of luminal progenitors. Nat Commun 2021; 12:1502. [PMID: 33686070 PMCID: PMC7940427 DOI: 10.1038/s41467-021-21783-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/11/2021] [Indexed: 12/13/2022] Open
Abstract
It is unclear how genetic aberrations impact the state of nascent tumour cells and their microenvironment. BRCA1 driven triple negative breast cancer (TNBC) has been shown to arise from luminal progenitors yet little is known about how BRCA1 loss-of-function (LOF) and concomitant mutations affect the luminal progenitor cell state. Here we demonstrate how time-resolved single-cell profiling of genetically engineered mouse models before tumour formation can address this challenge. We found that perturbing Brca1/p53 in luminal progenitors induces aberrant alveolar differentiation pre-malignancy accompanied by pro-tumourigenic changes in the immune compartment. Unlike alveolar differentiation during gestation, this process is cell autonomous and characterised by the dysregulation of transcription factors driving alveologenesis. Based on our data we propose a model where Brca1/p53 LOF inadvertently promotes a differentiation program hardwired in luminal progenitors, highlighting the deterministic role of the cell-of-origin and offering a potential explanation for the tissue specificity of BRCA1 tumours.
Collapse
Affiliation(s)
- Karsten Bach
- University of Cambridge, Department of Pharmacology, Cambridge, UK
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
- Cancer Research UK, Cambridge Cancer Centre, Cambridge, UK
| | - Sara Pensa
- University of Cambridge, Department of Pharmacology, Cambridge, UK
- Cancer Research UK, Cambridge Cancer Centre, Cambridge, UK
| | - Marija Zarocsinceva
- Cancer Research UK, Cambridge Cancer Centre, Cambridge, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
| | - Katarzyna Kania
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Julie Stockis
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Silvain Pinaud
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Kyren A Lazarus
- University of Cambridge, Department of Pharmacology, Cambridge, UK
- Cancer Research UK, Cambridge Cancer Centre, Cambridge, UK
| | - Mona Shehata
- Medical Research Council Cancer Unit, University of Cambridge, Cambridge, UK
| | - Bruno M Simões
- Manchester Breast Centre, Oglesby Cancer Research Building, University of Manchester, Manchester, UK
| | - Alice R Greenhalgh
- Manchester Breast Centre, Oglesby Cancer Research Building, University of Manchester, Manchester, UK
| | - Sacha J Howell
- Manchester Breast Centre, Oglesby Cancer Research Building, University of Manchester, Manchester, UK
- Department of Medical Oncology, Christie NHS Foundation Trust, Manchester, UK
| | - Robert B Clarke
- Manchester Breast Centre, Oglesby Cancer Research Building, University of Manchester, Manchester, UK
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
- Cancer Research UK, Cambridge Cancer Centre, Cambridge, UK
| | - Timotheus Y F Halim
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - John C Marioni
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK.
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.
- European Bioinformatics Institute, European Molecular Biology Laboratory, Hinxton, UK.
| | - Walid T Khaled
- University of Cambridge, Department of Pharmacology, Cambridge, UK.
- Cancer Research UK, Cambridge Cancer Centre, Cambridge, UK.
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK.
| |
Collapse
|
43
|
Dawson CA, Visvader JE. The Cellular Organization of the Mammary Gland: Insights From Microscopy. J Mammary Gland Biol Neoplasia 2021; 26:71-85. [PMID: 33835387 DOI: 10.1007/s10911-021-09483-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/25/2021] [Indexed: 12/19/2022] Open
Abstract
Despite rapid advances in our knowledge of the cellular heterogeneity and molecular regulation of the mammary gland, how these relate to 3D cellular organization remains unclear. In addition to hormonal regulation, mammary gland development and function is directed by para- and juxtacrine signaling among diverse cell-types, particularly the immune and mesenchymal populations. Precise mapping of the cellular landscape of the breast will help to decipher this complex coordination. Imaging of thin tissue sections has provided foundational information about cell positioning in the mammary gland and now technological advances in tissue clearing and subcellular-resolution 3D imaging are painting a more complete picture. In particular, confocal, light-sheet and multiphoton microscopy applied to intact tissue can fully capture cell morphology, position and interactions, and have the power to identify spatially rare events. This review will summarize our current understanding of mammary gland cellular organization as revealed by microscopy. We focus on the mouse mammary gland and cover a broad range of immune and stromal cell types at major developmental stages and give insights into important tissue niches and cellular interactions.
Collapse
Affiliation(s)
- Caleb A Dawson
- Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, 3052, Parkville, VIC, Australia.
- Department of Medical Biology, The University of Melbourne, 3010, Parkville, VIC, Australia.
| | - Jane E Visvader
- Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, 3052, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, 3010, Parkville, VIC, Australia
| |
Collapse
|
44
|
Neo WH, Lie-A-Ling M, Fadlullah MZH, Lacaud G. Contributions of Embryonic HSC-Independent Hematopoiesis to Organogenesis and the Adult Hematopoietic System. Front Cell Dev Biol 2021; 9:631699. [PMID: 33681211 PMCID: PMC7930747 DOI: 10.3389/fcell.2021.631699] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/22/2021] [Indexed: 12/17/2022] Open
Abstract
During ontogeny, the establishment of the hematopoietic system takes place in several phases, separated both in time and location. The process is initiated extra-embryonically in the yolk sac (YS) and concludes in the main arteries of the embryo with the formation of hematopoietic stem cells (HSC). Initially, it was thought that HSC-independent hematopoietic YS cells were transient, and only required to bridge the gap to HSC activity. However, in recent years it has become clear that these cells also contribute to embryonic organogenesis, including the emergence of HSCs. Furthermore, some of these early HSC-independent YS cells persist into adulthood as distinct hematopoietic populations. These previously unrecognized abilities of embryonic HSC-independent hematopoietic cells constitute a new field of interest. Here, we aim to provide a succinct overview of the current knowledge regarding the contribution of YS-derived hematopoietic cells to the development of the embryo and the adult hematopoietic system.
Collapse
Affiliation(s)
- Wen Hao Neo
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Macclesfield, United Kingdom
| | - Michael Lie-A-Ling
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Macclesfield, United Kingdom
| | | | - Georges Lacaud
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Macclesfield, United Kingdom
| |
Collapse
|
45
|
Bijnen M, Bajénoff M. Gland Macrophages: Reciprocal Control and Function within Their Niche. Trends Immunol 2021; 42:120-136. [PMID: 33423933 DOI: 10.1016/j.it.2020.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 11/30/2022]
Abstract
The human body contains dozens of endocrine and exocrine glands, which regulate physiological processes by secreting hormones and other factors. Glands can be subdivided into contiguous tissue modules, each consisting of an interdependent network of cells that together perform particular tissue functions. Among those cells are macrophages, a diverse type of immune cells endowed with trophic functions. In this review, we discuss recent findings on how resident macrophages support tissue modules within glands via the creation of mutually beneficial cell-cell circuits. A better comprehension of gland macrophage function and local control within their niche is essential to achieve a refined understanding of gland physiology in homeostasis and disease.
Collapse
Affiliation(s)
- Mitchell Bijnen
- Aix Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France.
| | - Marc Bajénoff
- Aix Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| |
Collapse
|
46
|
TIME Is a Great Healer-Targeting Myeloid Cells in the Tumor Immune Microenvironment to Improve Triple-Negative Breast Cancer Outcomes. Cells 2020; 10:cells10010011. [PMID: 33374595 PMCID: PMC7822423 DOI: 10.3390/cells10010011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/15/2020] [Accepted: 12/21/2020] [Indexed: 12/26/2022] Open
Abstract
The word myeloid is derived from the Greek word muelós which means "marrow". Therefore, myeloid cells are described as cells that arise in the bone marrow. They can be distinguished from lymphoid cells based on their different differentiation trajectories-Lymphoid cells (B and T cells) are usually born in the bone marrow, but they need to migrate to lymphoid organs to mature and differentiate usually in response to antigens produced due to infections and diseases like cancer. On the other hand, myeloid cells do not follow this differentiation trajectory. They arise from the bone marrow, and do not need an encounter with antigens to gain their functionality. Thus, while lymphoid cells are a part of the adaptive immune system, myeloid cells are a part of the innate immune system. Hematopoiesis gives rise to two progenitor cells-the common myeloid progenitor (CMP) and the common lymphoid progenitor (CLP). The CMP can give rise to megakaryocytes, erythrocytes, mast cells and myeloblasts. Myeloblasts in turn lead to the formation of basophils, neutrophils, eosinophils and monocytes that can further differentiate into macrophages. This review will focus on macrophages as well as the phenotypes they acquire with the tumor immune microenvironment (TIME) in triple-negative breast cancer (TNBC). It will address how cancer cells in the tumor microenvironment (TME) recruit macrophages and may switch to recruiting neutrophils upon depletion of these tumor-associated macrophages (TAMs). Finally, it will also shed light on past and current treatment options that specifically target these cells and how those affect patient outcomes in TNBC.
Collapse
|
47
|
Strack E, Rolfe PA, Fink AF, Bankov K, Schmid T, Solbach C, Savai R, Sha W, Pradel L, Hartmann S, Brüne B, Weigert A. Identification of tumor-associated macrophage subsets that are associated with breast cancer prognosis. Clin Transl Med 2020; 10:e239. [PMID: 33377644 PMCID: PMC7719284 DOI: 10.1002/ctm2.239] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/06/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Breast cancer is the leading cause of cancer-related deaths in women, demanding new treatment options. With the advent of immune checkpoint blockade, immunotherapy emerged as a treatment option. In addition to lymphocytes, tumor-associated macrophages exert a significant, albeit controversial, impact on tumor development. Pro-inflammatory macrophages are thought to hinder, whereas anti-inflammatory macrophages promote tumor growth. However, molecular markers to identify prognostic macrophage populations remain elusive. METHODS We isolated two macrophage subsets, from 48 primary human breast tumors, distinguished by the expression of CD206. Their transcriptomes were analyzed via RNA-Seq, and potential prognostic macrophage markers were validated by PhenOptics in tissue microarrays of patients with invasive breast cancer. RESULTS Normal human breast tissue contained mainly CD206+ macrophages, while increased relative amounts of CD206- macrophages were observed in tumors. The presence of CD206+ macrophages correlated with a pronounced lymphocyte infiltrate and subsets of CD206+ macrophages, expressing SERPINH1 and collagen 1, or MORC4, were unexpectedly associated with improved survival of breast cancer patients. In contrast, MHCIIhi CD206- macrophages were linked with a poor survival prognosis. CONCLUSION Our data highlight the heterogeneity of tumor-infiltrating macrophages and suggest the use of multiple phenotypic markers to predict the impact of macrophage subpopulations on cancer prognosis. We identified novel macrophage markers that correlate with the survival of patients with invasive mammary carcinoma.
Collapse
Affiliation(s)
- Elisabeth Strack
- Faculty of MedicineInstitute of Biochemistry IGoethe‐University FrankfurtFrankfurtGermany
| | | | - Annika F. Fink
- Faculty of MedicineInstitute of Biochemistry IGoethe‐University FrankfurtFrankfurtGermany
| | - Katrin Bankov
- Dr. Senckenberg Institute of PathologyUniversity Hospital FrankfurtFrankfurtGermany
| | - Tobias Schmid
- Faculty of MedicineInstitute of Biochemistry IGoethe‐University FrankfurtFrankfurtGermany
| | - Christine Solbach
- Department of GynecologyUniversity Hospital FrankfurtFrankfurtGermany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung ResearchBad NauheimGermany
- Frankfurt Cancer Institute (FCI)Goethe University FrankfurtFrankfurtGermany
- Institute of Lung Health (ILH)Justus Liebig UniversityGiessenGermany
| | | | | | - Sylvia Hartmann
- Dr. Senckenberg Institute of PathologyUniversity Hospital FrankfurtFrankfurtGermany
| | - Bernhard Brüne
- Faculty of MedicineInstitute of Biochemistry IGoethe‐University FrankfurtFrankfurtGermany
- Frankfurt Cancer Institute (FCI)Goethe University FrankfurtFrankfurtGermany
- German Cancer Consortium (DKTK)Partner Site FrankfurtFrankfurtGermany
| | - Andreas Weigert
- Faculty of MedicineInstitute of Biochemistry IGoethe‐University FrankfurtFrankfurtGermany
- Frankfurt Cancer Institute (FCI)Goethe University FrankfurtFrankfurtGermany
| |
Collapse
|
48
|
Aging-Associated Alterations in Mammary Epithelia and Stroma Revealed by Single-Cell RNA Sequencing. Cell Rep 2020; 33:108566. [PMID: 33378681 PMCID: PMC7898263 DOI: 10.1016/j.celrep.2020.108566] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/13/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
Aging is closely associated with increased susceptibility to breast cancer, yet there have been limited systematic studies of aging-induced alterations in the mammary gland. Here, we leverage high-throughput single-cell RNA sequencing to generate a detailed transcriptomic atlas of young and aged murine mammary tissues. By analyzing epithelial, stromal, and immune cells, we identify age-dependent alterations in cell proportions and gene expression, providing evidence that suggests alveolar maturation and physiological decline. The analysis also uncovers potential pro-tumorigenic mechanisms coupled to the age-associated loss of tumor suppressor function and change in microenvironment. In addition, we identify a rare, age-dependent luminal population co-expressing hormone-sensing and secretory-alveolar lineage markers, as well as two macrophage populations expressing distinct gene signatures, underscoring the complex heterogeneity of the mammary epithelia and stroma. Collectively, this rich single-cell atlas reveals the effects of aging on mammary physiology and can serve as a useful resource for understanding aging-associated cancer risk. Using single-cell RNA-sequencing, Li et al. compare mammary epithelia and stroma in young and aged mice. Age-dependent changes at cell and gene levels provide evidence suggesting alveolar maturation, functional deterioration, and potential pro-tumorigenic and inflammatory alterations. Additionally, identification of heterogeneous luminal and macrophage subpopulations underscores the complexity of mammary lineages.
Collapse
|
49
|
Etzerodt A, Moulin M, Doktor TK, Delfini M, Mossadegh-Keller N, Bajenoff M, Sieweke MH, Moestrup SK, Auphan-Anezin N, Lawrence T. Tissue-resident macrophages in omentum promote metastatic spread of ovarian cancer. J Exp Med 2020; 217:133611. [PMID: 31951251 PMCID: PMC7144521 DOI: 10.1084/jem.20191869] [Citation(s) in RCA: 193] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/23/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022] Open
Abstract
Experimental and clinical evidence suggests that tumor-associated macrophages (TAMs) play important roles in cancer progression. Here, we have characterized the ontogeny and function of TAM subsets in a mouse model of metastatic ovarian cancer that is representative for visceral peritoneal metastasis. We show that the omentum is a critical premetastatic niche for development of invasive disease in this model and define a unique subset of CD163+ Tim4+ resident omental macrophages responsible for metastatic spread of ovarian cancer cells. Transcriptomic analysis showed that resident CD163+ Tim4+ omental macrophages were phenotypically distinct and maintained their resident identity during tumor growth. Selective depletion of CD163+ Tim4+ macrophages in omentum using genetic and pharmacological tools prevented tumor progression and metastatic spread of disease. These studies describe a specific role for tissue-resident macrophages in the invasive progression of metastatic ovarian cancer. The molecular pathways of cross-talk between tissue-resident macrophages and disseminated cancer cells may represent new targets to prevent metastasis and disease recurrence.
Collapse
Affiliation(s)
- Anders Etzerodt
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.,Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - Morgane Moulin
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.,Centre for Inflammation Biology and Cancer Immunology, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Thomas Koed Doktor
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | | | | | - Marc Bajenoff
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Michael H Sieweke
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.,Centre for Regenerative Therapies, TU Dresden, Dresden, Germany
| | - Søren Kragh Moestrup
- Department of Biomedicine, University of Aarhus, Aarhus, Denmark.,Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Toby Lawrence
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.,Centre for Inflammation Biology and Cancer Immunology, School of Immunology & Microbial Sciences, King's College London, London, UK.,Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
50
|
Ibrahim AM, Moss MA, Gray Z, Rojo MD, Burke CM, Schwertfeger KL, Dos Santos CO, Machado HL. Diverse Macrophage Populations Contribute to the Inflammatory Microenvironment in Premalignant Lesions During Localized Invasion. Front Oncol 2020; 10:569985. [PMID: 33072601 PMCID: PMC7541939 DOI: 10.3389/fonc.2020.569985] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 08/13/2020] [Indexed: 12/14/2022] Open
Abstract
Myeloid cell heterogeneity remains poorly studied in breast cancer, and particularly in premalignancy. Here, we used single cell RNA sequencing to characterize macrophage diversity in mouse pre-invasive lesions as compared to lesions undergoing localized invasion. Several subpopulations of macrophages with transcriptionally distinct profiles were identified, two of which resembled macrophages in the steady state. While all subpopulations expressed tumor-promoting genes, many of the populations expressed pro-inflammatory genes, differing from reports in tumor-associated macrophages. Gene profiles of the myeloid cells were similar between early and late stages of premalignancy, although expansion of some subpopulations occurred. These results unravel macrophage heterogeneity in early progression and may provide insight into early intervention strategies that target macrophages.
Collapse
Affiliation(s)
- Ayman M Ibrahim
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, United States.,Tulane Cancer Center, Louisiana Cancer Research Consortium, New Orleans, LA, United States.,Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | - Matthew A Moss
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Zane Gray
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, United States.,Tulane Cancer Center, Louisiana Cancer Research Consortium, New Orleans, LA, United States
| | - Michelle D Rojo
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, United States.,Tulane Cancer Center, Louisiana Cancer Research Consortium, New Orleans, LA, United States
| | - Caitlin M Burke
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, United States.,Tulane Cancer Center, Louisiana Cancer Research Consortium, New Orleans, LA, United States
| | - Kathryn L Schwertfeger
- Department of Laboratory Medicine and Pathology, Masonic Cancer Center, and Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Camila O Dos Santos
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, United States
| | - Heather L Machado
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, United States.,Tulane Cancer Center, Louisiana Cancer Research Consortium, New Orleans, LA, United States
| |
Collapse
|