51
|
Nath PR, Gangaplara A, Pal-Nath D, Mandal A, Maric D, Sipes JM, Cam M, Shevach EM, Roberts DD. CD47 Expression in Natural Killer Cells Regulates Homeostasis and Modulates Immune Response to Lymphocytic Choriomeningitis Virus. Front Immunol 2018; 9:2985. [PMID: 30643501 PMCID: PMC6320676 DOI: 10.3389/fimmu.2018.02985] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 12/04/2018] [Indexed: 01/08/2023] Open
Abstract
CD47 is a ubiquitous cell surface receptor that directly regulates T cell immunity by interacting with its inhibitory ligand thrombospondin-1 and limits clearance of cells by phagocytes that express its counter-receptor signal-regulatory protein-α. Murine natural killer (NK) cells express higher levels of CD47 than other lymphocytes, but the role of CD47 in regulating NK cell homeostasis and immune function remains unclear. Cd47 -/- mice exhibited depletion of NK precursors in bone marrow, consistent with the antiphagocytic function of CD47. In contrast, antisense CD47 knockdown or gene disruption resulted in a dose dependent accumulation of immature and mature NK cells in spleen. Mature Cd47 -/- NK cells exhibited increased expression of NK effector and interferon gene signatures and an increased proliferative response to interleukin-15 in vitro. Cd47 -/- mice showed no defect in their early response to acute Armstrong lymphocytic choriomeningitis virus (LCMV) infection but were moderately impaired in controlling chronic Clone-13 LCMV infection, which was associated with depletion of splenic NK cells and loss of effector cytokine and interferon response gene expression in Cd47 -/- NK cells. Broad CD47-dependent differences in NK activation, survival, and exhaustion pathways were observed in NK cell transcriptional signatures in LCMV infected mice. These data identify CD47 as a cell-intrinsic and systemic regulator of NK cell homeostasis and NK cell function in responding to a viral infection.
Collapse
Affiliation(s)
- Pulak Ranjan Nath
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Arunakumar Gangaplara
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Dipasmita Pal-Nath
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ajeet Mandal
- CCR Collaborative Bioinformatics Resource, Office of Science and Technology Resources, National Cancer Institute and Leidos Biomedical Research, Inc., National Institutes of Health, Bethesda, MD, United States
| | - Dragan Maric
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - John M Sipes
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Maggie Cam
- CCR Collaborative Bioinformatics Resource, Office of Science and Technology Resources, National Cancer Institute and Leidos Biomedical Research, Inc., National Institutes of Health, Bethesda, MD, United States
| | - Ethan M Shevach
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
52
|
Pfeifhofer-Obermair C, Tymoszuk P, Petzer V, Weiss G, Nairz M. Iron in the Tumor Microenvironment-Connecting the Dots. Front Oncol 2018; 8:549. [PMID: 30534534 PMCID: PMC6275298 DOI: 10.3389/fonc.2018.00549] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/06/2018] [Indexed: 12/18/2022] Open
Abstract
Iron metabolism and tumor biology are intimately linked. Iron facilitates the production of oxygen radicals, which may either result in iron-induced cell death, ferroptosis, or contribute to mutagenicity and malignant transformation. Once transformed, malignant cells require high amounts of iron for proliferation. In addition, iron has multiple regulatory effects on the immune system, thus affecting tumor surveillance by immune cells. For these reasons, inconsiderate iron supplementation in cancer patients has the potential of worsening disease course and outcome. On the other hand, chronic immune activation in the setting of malignancy alters systemic iron homeostasis and directs iron fluxes into myeloid cells. While this response aims at withdrawing iron from tumor cells, it may impair the effector functions of tumor-associated macrophages and will result in iron-restricted erythropoiesis and the development of anemia, subsequently. This review summarizes our current knowledge of the interconnections of iron homeostasis with cancer biology, discusses current clinical controversies in the treatment of anemia of cancer and focuses on the potential roles of iron in the solid tumor microenvironment, also speculating on yet unknown molecular mechanisms.
Collapse
Affiliation(s)
- Christa Pfeifhofer-Obermair
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Piotr Tymoszuk
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Verena Petzer
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
53
|
Nemkov T, Reisz JA, Xia Y, Zimring JC, D’Alessandro A. Red blood cells as an organ? How deep omics characterization of the most abundant cell in the human body highlights other systemic metabolic functions beyond oxygen transport. Expert Rev Proteomics 2018; 15:855-864. [DOI: 10.1080/14789450.2018.1531710] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Aurora, CO, USA
| | - Julie A. Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Aurora, CO, USA
| | - Yang Xia
- Department of Biochemistry, University of Texas Houston – McGovern Medical School , Houston, TX, USA
| | | | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Aurora, CO, USA
| |
Collapse
|
54
|
Abstract
CD4+ T follicular helper (TFH) cells support germinal center (GC) reactions promoting humoral immunity. Dendritic cell (DC) diversification into genetically distinct subsets allows for specialization in promoting responses against several types of pathogens. Whether any classical DC (cDC) subset is required for humoral immunity is unknown, however. We tested several genetic models that selectively ablate distinct DC subsets in mice for their impact on splenic GC reactions. We identified a requirement for Notch2-dependent cDC2s, but not Batf3-dependent cDC1s or Klf4-dependent cDC2s, in promoting TFH and GC B cell formation in response to sheep red blood cells and inactivated Listeria monocytogenes This effect was mediated independent of Il2ra and several Notch2-dependent genes expressed in cDC2s, including Stat4 and Havcr2 Notch2 signaling during cDC2 development also substantially reduced the efficiency of cDC2s for presentation of MHC class II-restricted antigens, limiting the strength of CD4 T cell activation. Together, these results demonstrate a nonredundant role for the Notch2-dependent cDC2 subset in supporting humoral immune responses.
Collapse
|
55
|
Hong S, Zhang Z, Liu H, Tian M, Zhu X, Zhang Z, Wang W, Zhou X, Zhang F, Ge Q, Zhu B, Tang H, Hua Z, Hou B. B Cells Are the Dominant Antigen-Presenting Cells that Activate Naive CD4+ T Cells upon Immunization with a Virus-Derived Nanoparticle Antigen. Immunity 2018; 49:695-708.e4. [DOI: 10.1016/j.immuni.2018.08.012] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 03/27/2018] [Accepted: 08/07/2018] [Indexed: 12/23/2022]
|
56
|
Kauder SE, Kuo TC, Harrabi O, Chen A, Sangalang E, Doyle L, Rocha SS, Bollini S, Han B, Sim J, Pons J, Wan HI. ALX148 blocks CD47 and enhances innate and adaptive antitumor immunity with a favorable safety profile. PLoS One 2018; 13:e0201832. [PMID: 30133535 PMCID: PMC6104973 DOI: 10.1371/journal.pone.0201832] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/23/2018] [Indexed: 02/06/2023] Open
Abstract
CD47 is a widely expressed cell surface protein that functions as an immune checkpoint in cancer. When expressed by tumor cells, CD47 can bind SIRPα on myeloid cells, leading to suppression of tumor cell phagocytosis and other innate immune functions. CD47-SIRPα signaling has also been implicated in the suppression of adaptive antitumor responses, but the relevant cellular functions have yet to be elucidated. Therapeutic blockade of the CD47 pathway may stimulate antitumor immunity and improve cancer therapy. To this end, a novel CD47-blocking molecule, ALX148, was generated by fusing a modified SIRPα D1 domain to an inactive human IgG1 Fc. ALX148 binds CD47 from multiple species with high affinity, inhibits wild type SIRPα binding, and enhances phagocytosis of tumor cells by macrophages. ALX148 has no effect on normal human blood cells in vitro or on blood cell parameters in rodent and non-human primate studies. Across several murine tumor xenograft models, ALX148 enhanced the antitumor activity of different targeted antitumor antibodies. Additionally, ALX148 enhanced the antitumor activity of multiple immunotherapeutic antibodies in syngeneic tumor models. These studies revealed that CD47 blockade with ALX148 induces multiple responses that bridge innate and adaptive immunity. ALX148 stimulates antitumor properties of innate immune cells by promoting dendritic cell activation, macrophage phagocytosis, and a shift of tumor-associated macrophages toward an inflammatory phenotype. ALX148 also stimulated the antitumor properties of adaptive immune cells, causing increased T cell effector function, pro-inflammatory cytokine production, and a reduction in the number of suppressive cells within the tumor microenvironment. Taken together, these results show that ALX148 binds and blocks CD47 with high affinity, induces a broad antitumor immune response, and has a favorable safety profile.
Collapse
Affiliation(s)
| | - Tracy C. Kuo
- ALX Oncology, Burlingame, CA, United States of America
| | - Ons Harrabi
- ALX Oncology, Burlingame, CA, United States of America
| | - Amy Chen
- ALX Oncology, Burlingame, CA, United States of America
| | | | - Laura Doyle
- ALX Oncology, Burlingame, CA, United States of America
| | - Sony S. Rocha
- ALX Oncology, Burlingame, CA, United States of America
| | | | - Bora Han
- ALX Oncology, Burlingame, CA, United States of America
| | - Janet Sim
- ALX Oncology, Burlingame, CA, United States of America
| | - Jaume Pons
- ALX Oncology, Burlingame, CA, United States of America
| | - Hong I. Wan
- ALX Oncology, Burlingame, CA, United States of America
| |
Collapse
|
57
|
Krishnaswamy JK, Gowthaman U, Zhang B, Mattsson J, Szeponik L, Liu D, Wu R, White T, Calabro S, Xu L, Collet MA, Yurieva M, Alsén S, Fogelstrand P, Walter A, Heath WR, Mueller SN, Yrlid U, Williams A, Eisenbarth SC. Migratory CD11b + conventional dendritic cells induce T follicular helper cell-dependent antibody responses. Sci Immunol 2018; 2:2/18/eaam9169. [PMID: 29196450 DOI: 10.1126/sciimmunol.aam9169] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 08/11/2017] [Accepted: 10/19/2017] [Indexed: 12/18/2022]
Abstract
T follicular helper (Tfh) cells are a subset of CD4+ T cells that promote antibody production during vaccination. Conventional dendritic cells (cDCs) efficiently prime Tfh cells; however, conclusions regarding which cDC instructs Tfh cell differentiation have differed between recent studies. We found that these discrepancies might exist because of the unusual sites used for immunization in murine models, which differentially bias which DC subsets access antigen. We used intranasal immunization as a physiologically relevant route of exposure that delivers antigen to all tissue DC subsets. Using a combination of mice in which the function of individual DC subsets is impaired and different antigen formulations, we determined that CD11b+ migratory type 2 cDCs (cDC2s) are necessary and sufficient for Tfh induction. DC-specific deletion of the guanine nucleotide exchange factor DOCK8 resulted in an isolated loss of CD11b+ cDC2, but not CD103+ cDC1, migration to lung-draining lymph nodes. Impaired cDC2 migration or development in DC-specific Dock8 or Irf4 knockout mice, respectively, led to reduced Tfh cell priming, whereas loss of CD103+ cDC1s in Batf3-/- mice did not. Loss of cDC2-dependent Tfh cell priming impaired antibody-mediated protection from live influenza virus challenge. We show that migratory cDC2s uniquely carry antigen into the subanatomic regions of the lymph node where Tfh cell priming occurs-the T-B border. This work identifies the DC subset responsible for Tfh cell-dependent antibody responses, particularly when antigen dose is limiting or is encountered at a mucosal site, which could ultimately inform the formulation and delivery of vaccines.
Collapse
Affiliation(s)
- Jayendra Kumar Krishnaswamy
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA.,Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, 431 50 Mölndal, Sweden
| | - Uthaman Gowthaman
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Biyan Zhang
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Johan Mattsson
- Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, 431 50 Mölndal, Sweden
| | - Louis Szeponik
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Dong Liu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Renee Wu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.,Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Theresa White
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Samuele Calabro
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA.,Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Lan Xu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Magalie A Collet
- Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Marina Yurieva
- Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Samuel Alsén
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Per Fogelstrand
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Anne Walter
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, Victoria 3000, Australia
| | - William R Heath
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Ulf Yrlid
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Adam Williams
- Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA. .,Department of Genetics and Genomic Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA. .,Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
58
|
Nairz M, Dichtl S, Schroll A, Haschka D, Tymoszuk P, Theurl I, Weiss G. Iron and innate antimicrobial immunity-Depriving the pathogen, defending the host. J Trace Elem Med Biol 2018; 48:118-133. [PMID: 29773170 DOI: 10.1016/j.jtemb.2018.03.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/25/2018] [Accepted: 03/06/2018] [Indexed: 02/08/2023]
Abstract
The acute-phase response is triggered by the presence of infectious agents and danger signals which indicate hazards for the integrity of the mammalian body. One central feature of this response is the sequestration of iron into storage compartments including macrophages. This limits the availability of this essential nutrient for circulating pathogens, a host defence strategy known as 'nutritional immunity'. Iron metabolism and the immune response are intimately linked. In infections, the availability of iron affects both the efficacy of antimicrobial immune pathways and pathogen proliferation. However, host strategies to withhold iron from microbes vary according to the localization of pathogens: Infections with extracellular bacteria such as Staphylococcus aureus, Streptococcus, Klebsiella or Yersinia stimulate the expression of the iron-regulatory hormone hepcidin which targets the cellular iron-exporter ferroportin-1 causing its internalization and blockade of iron egress from absorptive enterocytes in the duodenum and iron-recycling macrophages. This mechanism disrupts both routes of iron delivery to the circulation, contributes to iron sequestration in the mononuclear phagocyte system and mediates the hypoferraemia of the acute phase response subsequently resulting in the development of anaemia of inflammation. When intracellular microbes are present, other strategies of microbial iron withdrawal are needed. For instance, in macrophages harbouring intracellular pathogens such as Chlamydia, Mycobacterium tuberculosis, Listeria monocytogenes or Salmonella Typhimurium, ferroportin-1-mediated iron export is turned on for the removal of iron from infected cells. This also leads to reduced iron availability for intra-macrophage pathogens which inhibits their growth and in parallel strengthens anti-microbial effector pathways of macrophages including the formation of inducible nitric oxide synthase and tumour necrosis factor. Iron plays a key role in infectious diseases both as modulator of the innate immune response and as nutrient for microbes. We need to gain a more comprehensive understanding of how the body can differentially respond to infection by extra- or intracellular pathogens. This knowledge may allow us to modulate mammalian iron homeostasis pharmaceutically and to target iron-acquisition systems of pathogens, thus enabling us to treat infections with novel strategies that act independent of established antimicrobials.
Collapse
Affiliation(s)
- Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria.
| | - Stefanie Dichtl
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - Andrea Schroll
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - David Haschka
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - Piotr Tymoszuk
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - Igor Theurl
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| |
Collapse
|
59
|
Wu J, Wu H, An J, Ballantyne CM, Cyster JG. Critical role of integrin CD11c in splenic dendritic cell capture of missing-self CD47 cells to induce adaptive immunity. Proc Natl Acad Sci U S A 2018; 115:6786-6791. [PMID: 29891680 PMCID: PMC6042080 DOI: 10.1073/pnas.1805542115] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
CD11c, also known as integrin alpha X, is the most widely used defining marker for dendritic cells (DCs). CD11c can bind complement iC3b and mediate phagocytosis in vitro, for which it is also referred to as complement receptor 4. However, the functions of this prominent marker protein in DCs, especially in vivo, remain poorly defined. Here, in the process of studying DC activation and immune responses induced by cells lacking self-CD47, we found that DC capture of CD47-deficient cells and DC activation was dependent on the integrin-signaling adaptor Talin1. Specifically, CD11c and its partner Itgb2 were required for DC capture of CD47-deficient cells. CD11b was not necessary for this process but could partially compensate in the absence of CD11c. Mice with DCs lacking Talin1, Itgb2, or CD11c were defective in supporting T-cell proliferation and differentiation induced by CD47-deficient cell associated antigen. These findings establish a critical role for CD11c in DC antigen uptake and activation in vivo. They may also contribute to understanding the functional mechanism of CD47-blockade therapies.
Collapse
Affiliation(s)
- Jiaxi Wu
- Department of Microbiology and Immunology, University of California, San Francisco, CA 94143
| | - Huaizhu Wu
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Jinping An
- Department of Microbiology and Immunology, University of California, San Francisco, CA 94143
| | - Christie M Ballantyne
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital and Baylor College of Medicine, Houston, TX 77030
| | - Jason G Cyster
- Department of Microbiology and Immunology, University of California, San Francisco, CA 94143;
- Howard Hughes Medical Institute, University of California, San Francisco, CA 94143
| |
Collapse
|
60
|
Loetsch C, Warren J, Laskowski A, Vazquez-Lombardi R, Jandl C, Langley DB, Christ D, Thorburn DR, Ryugo DK, Sprent J, Batten M, King C. Cytosolic Recognition of RNA Drives the Immune Response to Heterologous Erythrocytes. Cell Rep 2018; 21:1624-1638. [PMID: 29117566 DOI: 10.1016/j.celrep.2017.10.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 08/07/2017] [Accepted: 10/11/2017] [Indexed: 12/14/2022] Open
Abstract
The archetypal T cell-dependent antigen is sheep red blood cells (SRBCs), which have defined much of what we know about humoral immunity. Early studies using solubilized or sonicated SRBCs argued that the intact structure of SRBCs was important for optimal antibody responses. However, the reason for the requirement of intact SRBCs for the response to polyvalent protein antigen remained unknown. Here, we report that the immune response to SRBCs is driven by cytosolic recognition of SRBC RNA through the RIG-I-like receptor (RLR)-mitochondrial anti-viral signaling adaptor (MAVS) pathway. Following the uptake of SRBCs by antigen-presenting cells, the MAVS signaling complex governs the differentiation of both T follicular cells and antibody-producing B cells. Importantly, the involvement of the RLR-MAVS pathway precedes that of endosomal Toll-like receptor pathways, yet both are required for optimal effect.
Collapse
Affiliation(s)
- Claudia Loetsch
- Department of Immunology, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Department of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Joanna Warren
- Department of Immunology, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia
| | - Adrienne Laskowski
- Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Rd., Parkville, VIC 3052, Australia
| | - Rodrigo Vazquez-Lombardi
- Department of Immunology, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Department of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Christoph Jandl
- Department of Immunology, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Department of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - David B Langley
- Department of Immunology, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Department of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Daniel Christ
- Department of Immunology, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Department of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - David R Thorburn
- Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Rd., Parkville, VIC 3052, Australia; Department of Paediatrics, University of Melbourne, Parkville VIC 3010, Australia
| | - David K Ryugo
- Department of Immunology, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Department of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Jonathan Sprent
- Department of Immunology, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Department of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Marcel Batten
- Department of Immunology, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Department of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Cecile King
- Department of Immunology, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Department of Medicine, University of New South Wales, Sydney, NSW 2010, Australia.
| |
Collapse
|
61
|
Mener A, Arthur CM, Patel SR, Liu J, Hendrickson JE, Stowell SR. Complement Component 3 Negatively Regulates Antibody Response by Modulation of Red Blood Cell Antigen. Front Immunol 2018; 9:676. [PMID: 29942300 PMCID: PMC6004516 DOI: 10.3389/fimmu.2018.00676] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 03/19/2018] [Indexed: 12/17/2022] Open
Abstract
Red blood cell (RBC) alloimmunization can make it difficult to procure compatible RBCs for future transfusion, directly leading to increased morbidity and mortality in transfusion-dependent patients. However, the factors that regulate RBC alloimmunization remain incompletely understood. As complement has been shown to serve as a key adjuvant in the development of antibody (Ab) responses against microbes, we examined the impact of complement on RBC alloimmunization. In contrast to the impact of complement component 3 (C3) in the development of an immune response following microbial exposure, transfusion of C3 knockout (C3 KO) recipients with RBCs expressing KEL (KEL RBCs) actually resulted in an enhanced anti-KEL Ab response. The impact of C3 appeared to be specific to KEL, as transfusion of RBCs bearing another model antigen, the chimeric HOD antigen (hen egg lysozyme, ovalbumin and Duffy), into C3 KO recipients failed to result in a similar increase in Ab formation. KEL RBCs experienced enhanced C3 deposition and loss of detectable target antigen over time when compared to HOD RBCs, suggesting that C3 may inhibit Ab formation by impacting the accessibility of the target KEL antigen. Loss of detectable KEL on the RBC surface did not reflect antigen masking by C3, but instead appeared to result from actual removal of the KEL antigen, as western blot analysis demonstrated complete loss of detectable KEL protein. Consistent with this, exposure of wild-type B6 or C3 KO recipients to KEL RBCs with reduced levels of detectable KEL antigen resulted in a significantly reduced anti-KEL Ab response. These results suggest that C3 possesses a unique ability to actually suppress Ab formation following transfusion by reducing the availability of the target antigen on the RBC surface.
Collapse
Affiliation(s)
- Amanda Mener
- Center for Transfusion Medicine and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Connie M Arthur
- Center for Transfusion Medicine and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Seema R Patel
- Center for Transfusion Medicine and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Jingchun Liu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Jeanne E Hendrickson
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Sean R Stowell
- Center for Transfusion Medicine and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
62
|
Murata Y, Tanaka D, Hazama D, Yanagita T, Saito Y, Kotani T, Oldenborg PA, Matozaki T. Anti-human SIRPα antibody is a new tool for cancer immunotherapy. Cancer Sci 2018; 109:1300-1308. [PMID: 29473266 PMCID: PMC5980332 DOI: 10.1111/cas.13548] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/17/2018] [Accepted: 02/19/2018] [Indexed: 01/15/2023] Open
Abstract
Interaction of signal regulatory protein α (SIRPα) expressed on the surface of macrophages with its ligand CD47 expressed on target cells negatively regulates phagocytosis of the latter cells by the former. We recently showed that blocking Abs to mouse SIRPα enhanced both the Ab‐dependent cellular phagocytosis (ADCP) activity of mouse macrophages for Burkitt's lymphoma Raji cells opsonized with an Ab to CD20 (rituximab) in vitro as well as the inhibitory effect of rituximab on the growth of tumors formed by Raji cells in nonobese diabetic (NOD)/SCID mice. However, the effects of blocking Abs to human SIRPα in preclinical cancer models have remained unclear given that such Abs have failed to interact with endogenous SIRPα expressed on macrophages of immunodeficient mice. With the use of Rag2−/−γc−/− mice harboring a transgene for human SIRPα under the control of human regulatory elements (hSIRPα‐DKO mice), we here show that a blocking Ab to human SIRPα significantly enhanced the ADCP activity of macrophages derived from these mice for human cancer cells. The anti‐human SIRPα Ab also markedly enhanced the inhibitory effect of rituximab on the growth of tumors formed by Raji cells in hSIRPα‐DKO mice. Our results thus suggest that the combination of Abs to human SIRPα with therapeutic Abs specific for tumor antigens warrants further investigation for potential application to cancer immunotherapy. In addition, humanized mice, such as hSIRPα‐DKO mice, should prove useful for validation of the antitumor effects of checkpoint inhibitors before testing in clinical trials.
Collapse
Affiliation(s)
- Yoji Murata
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Daisuke Tanaka
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Daisuke Hazama
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tadahiko Yanagita
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yasuyuki Saito
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takenori Kotani
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Per-Arne Oldenborg
- Department of Integrative Medical Biology, Section for Histology and Cell Biology, Umeå University, Umeå, Sweden
| | - Takashi Matozaki
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
63
|
Barnes MJ, Cyster JG. Lysophosphatidylserine suppression of T-cell activation via GPR174 requires Gαs proteins. Immunol Cell Biol 2018; 96:439-445. [PMID: 29457279 PMCID: PMC5916342 DOI: 10.1111/imcb.12025] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 02/13/2018] [Accepted: 02/13/2018] [Indexed: 01/26/2023]
Abstract
G protein-coupled receptors regulate diverse aspects of T-cell activity and effector function. Recently, we showed that GPR174 mediates the suppression of T-cell proliferation in vitro induced by the polar lipid lysophosphatidylserine (LysoPS). Here, we investigated the in vivo activity of this pathway and characterized the mechanisms involved. Using in vivo models of T-cell proliferation induced by sublethal irradiation or regulatory T-cell depletion, we show that GPR174 expression can constrain T-cell proliferation. In vitro experiments established that Gαs G proteins are needed for LysoPS/GPR174-mediated suppression of T-cell proliferation. Mechanistically, LysoPS acts via GPR174 and Gαs to suppress IL-2 production by activated T cells and limit upregulation of the activation markers CD25 and CD69. Together, our findings identify GPR174 as an abundantly expressed Gαs-dependent receptor that can negatively regulate naive T-cell activation. See also: News and Commentary by Robert & Mackay.
Collapse
Affiliation(s)
- Michael J. Barnes
- Howard Hughes Medical Institute and University of California, San Francisco; Department of Microbiology and Immunology; 513 Parnassus Ave. HSE-1001H; San Francisco, CA 94143, USA
| | - Jason G. Cyster
- Howard Hughes Medical Institute and University of California, San Francisco; Department of Microbiology and Immunology; 513 Parnassus Ave. HSE-1001H; San Francisco, CA 94143, USA
| |
Collapse
|
64
|
Dheilly E, Majocchi S, Moine V, Didelot G, Broyer L, Calloud S, Malinge P, Chatel L, Ferlin WG, Kosco-Vilbois MH, Fischer N, Masternak K. Tumor-Directed Blockade of CD47 with Bispecific Antibodies Induces Adaptive Antitumor Immunity. Antibodies (Basel) 2018; 7:antib7010003. [PMID: 31544856 PMCID: PMC6698848 DOI: 10.3390/antib7010003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/22/2017] [Accepted: 12/22/2017] [Indexed: 01/02/2023] Open
Abstract
CD47 serves as an anti-phagocytic receptor that is upregulated by cancer to promote immune escape. As such, CD47 is the focus of intense immuno-oncology drug development efforts. However, as CD47 is expressed ubiquitously, clinical development of conventional drugs, e.g., monoclonal antibodies, is confronted with patient safety issues and poor pharmacology due to the widespread CD47 “antigen sink”. A potential solution is tumor-directed blockade of CD47, which can be achieved with bispecific antibodies (biAbs). Using mouse CD47-blocking biAbs in a syngeneic tumor model allowed us to evaluate the efficacy of tumor-directed blockade of CD47 in the presence of the CD47 antigen sink and a functional adaptive immune system. We show here that CD47-targeting biAbs inhibited tumor growth in vivo, promoting durable antitumor responses and stimulating CD8+ T cell activation in vitro. In vivo efficacy of the biAbs could be further enhanced when combined with chemotherapy or PD-1/PD-L1 immune checkpoint blockade. We also show that selectivity and pharmacological properties of the biAb are dependent on the affinity of the anti-CD47 arm. Taken together, our study validates the approach to use CD47-blocking biAbs either as a monotherapy or part of a multi-drug approach to enhance antitumor immunity.
Collapse
Affiliation(s)
- Elie Dheilly
- Novimmune S.A., 14 chemin des Aulx, CH-1228 Geneva, Switzerland.
| | - Stefano Majocchi
- Novimmune S.A., 14 chemin des Aulx, CH-1228 Geneva, Switzerland.
| | - Valéry Moine
- Novimmune S.A., 14 chemin des Aulx, CH-1228 Geneva, Switzerland.
| | - Gérard Didelot
- Novimmune S.A., 14 chemin des Aulx, CH-1228 Geneva, Switzerland.
| | - Lucile Broyer
- Novimmune S.A., 14 chemin des Aulx, CH-1228 Geneva, Switzerland.
| | | | - Pauline Malinge
- Novimmune S.A., 14 chemin des Aulx, CH-1228 Geneva, Switzerland.
| | - Laurence Chatel
- Novimmune S.A., 14 chemin des Aulx, CH-1228 Geneva, Switzerland.
| | - Walter G Ferlin
- Novimmune S.A., 14 chemin des Aulx, CH-1228 Geneva, Switzerland.
| | | | - Nicolas Fischer
- Novimmune S.A., 14 chemin des Aulx, CH-1228 Geneva, Switzerland.
| | | |
Collapse
|
65
|
Spiller F, Nycholat CM, Kikuchi C, Paulson JC, Macauley MS. Murine Red Blood Cells Lack Ligands for B Cell Siglecs, Allowing Strong Activation by Erythrocyte Surface Antigens. THE JOURNAL OF IMMUNOLOGY 2017; 200:949-956. [PMID: 29288201 DOI: 10.4049/jimmunol.1701257] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/27/2017] [Indexed: 01/01/2023]
Abstract
CD22 and sialic acid-binding Ig-like lectin (Siglec)-G are members of the Siglec family of inhibitory coreceptors expressed on B cells that participate in enforcement of peripheral B cell tolerance. We have shown previously that when a BCR engages its cognate Ag on a cell surface that also expresses Siglec ligands, B cell Siglecs are recruited to the immunological synapse, resulting in suppression of BCR signaling and B cell apoptosis. Because all cells display sialic acids, and CD22 and Siglec-G have distinct, yet overlapping, specificities for sialic acid-containing glycan ligands, any cell could, in principle, invoke this tolerogenic mechanism for cell surface Ags. However, we show in this article that C57BL/6J mouse RBCs are essentially devoid of CD22 and Siglec-G ligands. As a consequence, RBCs that display a cell surface Ag, membrane-bound hen egg lysozyme, strongly activate Ag-specific B cells. We reasoned that de novo introduction of CD22 ligands in RBCs should abolish B cell activation toward its cognate Ag on the surface of RBCs. Accordingly, we used a glyco-engineering approach wherein synthetic CD22 ligands linked to lipids are inserted into the membrane of RBCs. Indeed, insertion of CD22 ligands into the RBC cell surface strongly inhibited B cell activation, cytokine secretion, and proliferation. These results demonstrate that the lack of Siglec ligands on the surface of murine RBCs permits B cell responses to erythrocyte Ags and show that Siglec-mediated B cell tolerance is restricted to cell types that express glycan ligands for the B cell Siglecs.
Collapse
Affiliation(s)
- Fernando Spiller
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037.,Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037; and
| | - Corwin M Nycholat
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037.,Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037; and
| | - Chika Kikuchi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037.,Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037; and
| | - James C Paulson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037; .,Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037; and
| | - Matthew S Macauley
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
66
|
de Souza HADS, Costa-Correa EH, Bianco-Junior C, Andrade MCR, Lima-Junior JDC, Pratt-Riccio LR, Daniel-Ribeiro CT, Totino PRR. Detection of Signal Regulatory Protein α in Saimiri sciureus (Squirrel Monkey) by Anti-Human Monoclonal Antibody. Front Immunol 2017; 8:1814. [PMID: 29312325 PMCID: PMC5735064 DOI: 10.3389/fimmu.2017.01814] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/01/2017] [Indexed: 12/18/2022] Open
Abstract
Non-human primates (NHP) are suitable models for studying different aspects of the human system, including pathogenesis and protective immunity to many diseases. However, the lack of specific immunological reagents for neo-tropical monkeys, such as Saimiri sciureus, is still a major factor limiting studies in these models. An alternative strategy to circumvent this obstacle has been the selection of immunological reagents directed to humans, which present cross-reactivity with NHP molecules. In this context and considering the key role of inhibitory immunoreceptors—such as the signal regulatory protein α (SIRPα)—in the regulation of immune responses, in the present study, we attempted to evaluate the ability of anti-human SIRPα monoclonal antibodies to recognize SIRPα in antigen-presenting S. sciureus peripheral blood mononuclear cells (PBMC). As shown by flow cytometry analysis, the profile of anti-SIRPα staining as well as the levels of SIRPα-positive cells in PBMC from S. sciureus were similar to those observed in human PBMC. Furthermore, using anti-SIRPα monoclonal antibody, it was possible to detect a decrease of the SIRPα levels on surface of S. sciureus cells after in vitro stimulation with lipopolysaccharides. Finally, using computed-based analysis, we observed a high degree of conservation of SIRPα across six species of primates and the presence of shared epitopes in the extracellular domain between humans and Saimiri genus that could be targeted by antibodies. In conclusion, we have identified a commercially available anti-human monoclonal antibody that is able to detect SIRPα of S. sciureus monkeys and that, therefore, can facilitate the study of the immunomodulatory role of SIRPα when S. sciureus is used as a model.
Collapse
Affiliation(s)
| | - Edmar Henrique Costa-Correa
- Laboratory for Malaria Research, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Cesare Bianco-Junior
- Laboratory for Malaria Research, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | | | | | - Lilian Rose Pratt-Riccio
- Laboratory for Malaria Research, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Cláudio Tadeu Daniel-Ribeiro
- Laboratory for Malaria Research, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Paulo Renato Rivas Totino
- Laboratory for Malaria Research, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| |
Collapse
|
67
|
SIRPα + dendritic cells regulate homeostasis of fibroblastic reticular cells via TNF receptor ligands in the adult spleen. Proc Natl Acad Sci U S A 2017; 114:E10151-E10160. [PMID: 29109283 DOI: 10.1073/pnas.1711345114] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In secondary lymphoid organs, development and homeostasis of stromal cells such as podoplanin (Pdpn)-positive fibroblastic reticular cells (FRCs) are regulated by hematopoietic cells, but the cellular and molecular mechanisms of such regulation have remained unclear. Here we show that ablation of either signal regulatory protein α (SIRPα), an Ig superfamily protein, or its ligand CD47 in conventional dendritic cells (cDCs) markedly reduced the number of CD4+ cDCs as well as that of Pdpn+ FRCs and T cells in the adult mouse spleen. Such ablation also impaired the survival of FRCs as well as the production by CD4+ cDCs of tumor necrosis factor receptor (TNFR) ligands, including TNF-α, which was shown to promote the proliferation and survival of Pdpn+ FRCs. CD4+ cDCs thus regulate the steady-state homeostasis of FRCs in the adult spleen via the production of TNFR ligands, with the CD47-SIRPα interaction in cDCs likely being indispensable for such regulation.
Collapse
|
68
|
Red blood cell alloimmunization: new findings at the bench and new recommendations for the bedside. Curr Opin Hematol 2017; 23:543-549. [PMID: 27454234 DOI: 10.1097/moh.0000000000000277] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
PURPOSE OF REVIEW To summarize recent discoveries from clinical studies and animal models that contribute to understanding the alloimmune response to non-ABO blood group antigens. RECENT FINDINGS Several studies have confirmed high rates of alloimmunization among patients requiring chronic red blood cell (RBC) transfusion. Moreover, 'triggers' for alloantibody development in the transfusion setting have been identified, with a number of investigations linking recipient inflammation to a higher likelihood of alloimmunization. Additional associations between human leukocyte antigen expression and CD4 T-cell markers in 'responder' or 'nonresponder' humans have been revealed. Recent animal studies have described novel mechanistic properties by which the alloimmune response is governed, including the critical role played by dendritic cells in transfusion-associated alloimmunization. New light has also been shed on the properties of alloantibodies developed as a result of pregnancy, as well as mechanisms through which such alloimmunization may be prevented. SUMMARY Many of the clinical/biological factors that contribute to the RBC alloimmune response have been further elucidated. This knowledge will be applied to identify individuals most likely to mount an immune response to RBC antigens, such that appropriate resources and strategies for preventing alloimmunization (or mitigating its harmful effects) can be implemented.
Collapse
|
69
|
Hendrickson JE. Red blood cell alloimmunisation: induction of immunity and potential mitigation strategies. ACTA ACUST UNITED AC 2017. [DOI: 10.1111/voxs.12360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- J. E. Hendrickson
- Department of Laboratory Medicine; Yale University School of Medicine; New Haven CT USA
- Department of Pediatrics; Yale University School of Medicine; New Haven CT USA
| |
Collapse
|
70
|
Matlung HL, Szilagyi K, Barclay NA, van den Berg TK. The CD47-SIRPα signaling axis as an innate immune checkpoint in cancer. Immunol Rev 2017; 276:145-164. [PMID: 28258703 DOI: 10.1111/imr.12527] [Citation(s) in RCA: 372] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Immune checkpoint inhibitors, including those targeting CTLA-4/B7 and the PD-1/PD-L1 inhibitory pathways, are now available for clinical use in cancer patients, with other interesting checkpoint inhibitors being currently in development. Most of these have the purpose to promote adaptive T cell-mediated immunity against cancer. Here, we review another checkpoint acting to potentiate the activity of innate immune cells towards cancer. This innate immune checkpoint is composed of what has become known as the 'don't-eat me' signal CD47, which is a protein broadly expressed on normal cells and often overexpressed on cancer cells, and its counter-receptor, the myeloid inhibitory immunoreceptor SIRPα. Blocking CD47-SIRPα interactions has been shown to promote the destruction of cancer cells by phagocytes, including macrophages and neutrophils. Furthermore, there is growing evidence that targeting of the CD47-SIRPα axis may also promote antigen-presenting cell function and thereby stimulate adaptive T cell-mediated anti-cancer immunity. The development of CD47-SIRPα checkpoint inhibitors and the potential side effects that these may have are discussed. Collectively, this identifies the CD47-SIRPα axis as a promising innate immune checkpoint in cancer, and with data of the first clinical studies with CD47-SIRPα checkpoint inhibitors expected within the coming years, this is an exciting and rapidly developing field.
Collapse
Affiliation(s)
- Hanke L Matlung
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Katka Szilagyi
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Neil A Barclay
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Timo K van den Berg
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Cell Biology and Immunology, VU medical Center, Amsterdam, The Netherlands
| |
Collapse
|
71
|
Dai H, Friday AJ, Abou-Daya KI, Williams AL, Mortin-Toth S, Nicotra ML, Rothstein DM, Shlomchik WD, Matozaki T, Isenberg JS, Oberbarnscheidt MH, Danska JS, Lakkis FG. Donor SIRPα polymorphism modulates the innate immune response to allogeneic grafts. Sci Immunol 2017; 2:2/12/eaam6202. [PMID: 28783664 DOI: 10.1126/sciimmunol.aam6202] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 05/15/2017] [Indexed: 12/16/2022]
Abstract
Mice devoid of T, B, and natural killer (NK) cells distinguish between self and allogeneic nonself despite the absence of an adaptive immune system. When challenged with an allograft, they mount an innate response characterized by accumulation of mature, monocyte-derived dendritic cells (DCs) that produce interleukin-12 and present antigen to T cells. However, the molecular mechanisms by which the innate immune system detects allogeneic nonself to generate these DCs are not known. To address this question, we studied the innate response of Rag2-/- γc-/- mice, which lack T, B, and NK cells, to grafts from allogeneic donors. By positional cloning, we identified that donor polymorphism in the gene encoding signal regulatory protein α (SIRPα) is a key modulator of the recipient's innate allorecognition response. Donors that differed from the recipient in one or both Sirpa alleles elicited an innate alloresponse. The response was mediated by binding of donor SIRPα to recipient CD47 and was modulated by the strength of the SIRPα-CD47 interaction. Therefore, sensing SIRPα polymorphism by CD47 provides a molecular mechanism by which the innate immune system distinguishes between self and allogeneic nonself independently of T, B, and NK cells.
Collapse
Affiliation(s)
- Hehua Dai
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Andrew J Friday
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Khodor I Abou-Daya
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Amanda L Williams
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Steven Mortin-Toth
- Program in Genetics and Genome Biology, Hospital for Sick Children Research Institute, Toronto, Ontario M5G1X8, Canada
| | - Matthew L Nicotra
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - David M Rothstein
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.,Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Warren D Shlomchik
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.,Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Takashi Matozaki
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Kobe 650-0017, Japan
| | - Jeffrey S Isenberg
- Heart, Lung, Blood, and Vascular Medicine Institute and Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Martin H Oberbarnscheidt
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.,Program in Genetics and Genome Biology, Hospital for Sick Children Research Institute, Toronto, Ontario M5G1X8, Canada
| | - Jayne S Danska
- Program in Genetics and Genome Biology, Hospital for Sick Children Research Institute, Toronto, Ontario M5G1X8, Canada. .,Departments of Immunology and Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Fadi G Lakkis
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA. .,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.,Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
72
|
Østergaard O, Nielsen CT, Tanassi JT, Iversen LV, Jacobsen S, Heegaard NHH. Distinct proteome pathology of circulating microparticles in systemic lupus erythematosus. Clin Proteomics 2017. [PMID: 28649187 PMCID: PMC5479039 DOI: 10.1186/s12014-017-9159-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background The pathogenesis of systemic lupus erythematosus (SLE) is poorly understood but has been linked to defective clearance of subcellular particulate material from the circulation. This study investigates the origin, formation, and specificity of circulating microparticles (MPs) in patients with SLE based on comprehensive MP proteome profiling using patients with systemic sclerosis (SSc) and healthy donors (HC) as controls. Methods We purified MPs from platelet-poor plasma using differential centrifugation of samples from SLE (n = 45), SSc (n = 38), and two sets of HC (n = 35, n = 25). MP proteins were identified and quantitated after trypsin digestion by liquid chromatography-tandem mass spectrometry. The abundance of specific proteins was compared between the groups using univariate statistics and false discovery rate correction for multiple comparisons. Specific proteins and protein ratios were explored for diagnostic and disease activity information using receiver-operating characteristic curves and by analysis of correlations of protein abundance with disease activity scores. Results We identify and quantitate more than 1000 MP proteins and show that a subpopulation of SLE-MPs (which we propose to call luposomes) are highly specific for SLE, i.e. not found in MP preparations from HC or patients with another autoimmune, systemic disease, SSc. In SLE-MPs platelet proteins and mitochondrial proteins are significantly diminished, cytoskeletal proteins deranged, and glycolytic enzymes and apoptotic proteins significantly increased. Conclusions Normal MPs are efficiently removed in SLE, but aberrant MPs, derived from non-lymphoid leukocytes, are less efficiently removed and abundantly produced leading to an altered MP proteome in SLE. The data suggest that an abnormal generation of MPs may partake in the pathology of SLE and that new diagnostic, monitoring, and treatment strategies targeting these processes may be advantageous. Electronic supplementary material The online version of this article (doi:10.1186/s12014-017-9159-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ole Østergaard
- Department of Autoimmunology and Biomarkers, Statens Serum Institut, Copenhagen, Denmark
| | - Christoffer Tandrup Nielsen
- Copenhagen Lupus and Vasculitis Clinic, Centre for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Julia T Tanassi
- Department of Autoimmunology and Biomarkers, Statens Serum Institut, Copenhagen, Denmark
| | - Line V Iversen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Søren Jacobsen
- Copenhagen Lupus and Vasculitis Clinic, Centre for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Niels H H Heegaard
- Department of Autoimmunology and Biomarkers, Statens Serum Institut, Copenhagen, Denmark.,Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
73
|
Burnett DL, Parish IA, Masle-Farquhar E, Brink R, Goodnow CC. Murine LRBA deficiency causes CTLA-4 deficiency in Tregs without progression to immune dysregulation. Immunol Cell Biol 2017; 95:775-788. [PMID: 28611475 PMCID: PMC5636941 DOI: 10.1038/icb.2017.50] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 06/06/2017] [Accepted: 06/06/2017] [Indexed: 12/12/2022]
Abstract
Inherited mutations in lipopolysaccharide-responsive beige-like anchor (LRBA) cause a recessive human immune dysregulation syndrome with memory B-cell and antibody deficiency (common variable immunodeficiency), inflammatory bowel disease, enlarged spleen and lymph nodes, accumulation of activated T cells and multiple autoimmune diseases. To understand the pathogenesis of the syndrome, C57BL/6 mice carrying a homozygous truncating mutation in Lrba were produced using CRISPR/Cas9-mediated gene targeting. These mice revealed that LRBA has a critical, cell-autonomous role in promoting cytotoxic T-lymphocyte antigen-4 (CTLA-4) accumulation within CD4 effector T cells and FOXP3+ T-regulatory cells (Tregs). In young mice, or in chimeric mice where only half of the T cells are LRBA deficient, low CTLA-4 was the only detectable abnormality in Tregs, whereas in old mice FOXP3 was also decreased. Low CTLA-4 did not translate into increased CD86 on B cells unless the LRBA-deficient mice were immunised, and neither immunisation nor chronic lymphocytic choriomeningitis virus infection precipitated immune dysregulation. LRBA deficiency did not alter antigen-specific B-cell activation, germinal centre (GC) formation, isotype switching or affinity maturation. Paradoxically, CD86 was decreased on GC B cells in LRBA-deficient mice, pointing to compensatory mechanisms for controlling CD86 in the face of low CTLA-4. These results add to the experimental rationale for treating LRBA deficiency with the CTLA4-Ig fusion protein, Abatacept, and pose questions about the limitations of laboratory experiments in mice to reproduce human disease in natura.
Collapse
Affiliation(s)
- Deborah L Burnett
- Immunology Division, Garvan Institute for Medical Research, Sydney, NSW, Australia
| | - Ian A Parish
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | | | - Robert Brink
- Immunology Division, Garvan Institute for Medical Research, Sydney, NSW, Australia
| | | |
Collapse
|
74
|
Lu E, Dang EV, McDonald JG, Cyster JG. Distinct oxysterol requirements for positioning naïve and activated dendritic cells in the spleen. Sci Immunol 2017; 2:2/10/eaal5237. [PMID: 28738017 DOI: 10.1126/sciimmunol.aal5237] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 02/17/2017] [Indexed: 12/15/2022]
Abstract
Correct positioning of dendritic cells (DCs) is critical for efficient pathogen encounter and antigen presentation. Epstein-Barr virus-induced gene 2 (EBI2) has been identified as a chemoattractant receptor required for naïve CD4+DCIR2+ DC positioning in response to 7α,25-hydroxycholesterol (7α,25-HC). We now provide evidence that a second EBI2 ligand, 7α,27-HC, is involved in splenic DCIR2+ DC positioning and homeostasis. Cyp27a1, the enzyme uniquely required for 7α,27-HC synthesis, is expressed by stromal cells in the region of naïve DC localization. After activation, DCIR2+ DCs move into the T cell zone. We find that EBI2 is rapidly up-regulated in DCIR2+ DCs under certain activation conditions, and positioning at the B-T zone interface depends on EBI2. Under conditions of type I interferon induction, EBI2 ligand levels are elevated, causing activated DCIR2+ DCs to disperse throughout the T zone. Last, we provide evidence that oxysterol metabolism by Batf3-dependent DCs is important for EBI2-dependent positioning of activated DCIR2+ DCs. This work indicates that 7α,27-HC functions as a guidance cue in vivo and reveals a multitiered role for EBI2 in DC positioning. Deficiency in this organizing system results in defective CD4+ T cell responses.
Collapse
Affiliation(s)
- Erick Lu
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California San Francisco, San Francisco, CA 94143, USA
| | - Eric V Dang
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jeffrey G McDonald
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jason G Cyster
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
75
|
Arias CF, Arias CF. How do red blood cells know when to die? ROYAL SOCIETY OPEN SCIENCE 2017; 4:160850. [PMID: 28484605 PMCID: PMC5414242 DOI: 10.1098/rsos.160850] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 03/01/2017] [Indexed: 05/19/2023]
Abstract
Human red blood cells (RBCs) are normally phagocytized by macrophages of splenic and hepatic sinusoids at 120 days of age. The destruction of RBCs is ultimately controlled by antagonist effects of phosphatidylserine (PS) and CD47 on the phagocytic activity of macrophages. In this work, we introduce a conceptual model that explains RBC lifespan as a consequence of the dynamics of these molecules. Specifically, we suggest that PS and CD47 define a molecular algorithm that sets the timing of RBC phagocytosis. We show that significant changes in RBC lifespan described in the literature can be explained as alternative outcomes of this algorithm when it is executed in different conditions of oxygen availability. The theoretical model introduced here provides a unified framework to understand a variety of empirical observations regarding RBC biology. It also highlights the role of RBC lifespan as a key element of RBC homeostasis.
Collapse
Affiliation(s)
- Clemente Fernandez Arias
- Departamento de Matemática Aplicada, Universidad Complutense de Madrid, Spain
- Grupo Interdisciplinar de Sistemas Complejos, Madrid, Spain
| | - Cristina Fernandez Arias
- HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY, USA
| |
Collapse
|
76
|
Weiskopf K. Cancer immunotherapy targeting the CD47/SIRPα axis. Eur J Cancer 2017; 76:100-109. [PMID: 28286286 DOI: 10.1016/j.ejca.2017.02.013] [Citation(s) in RCA: 243] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 01/29/2017] [Accepted: 02/05/2017] [Indexed: 12/31/2022]
Abstract
The success of cancer immunotherapy has generated tremendous interest in identifying new immunotherapeutic targets. To date, the majority of therapies have focussed on stimulating the adaptive immune system to attack cancer, including agents targeting CTLA-4 and the PD-1/PD-L1 axis. However, macrophages and other myeloid immune cells offer much promise as effectors of cancer immunotherapy. The CD47/signal regulatory protein alpha (SIRPα) axis is a critical regulator of myeloid cell activation and serves a broader role as a myeloid-specific immune checkpoint. CD47 is highly expressed on many different types of cancer, and it transduces inhibitory signals through SIRPα on macrophages and other myeloid cells. In a diverse range of preclinical models, therapies that block the CD47/SIRPα axis stimulate phagocytosis of cancer cells in vitro and anti-tumour immune responses in vivo. A number of therapeutics that target the CD47/SIRPα axis are under preclinical and clinical investigation. These include anti-CD47 antibodies, engineered receptor decoys, anti-SIRPα antibodies and bispecific agents. These therapeutics differ in their pharmacodynamic, pharmacokinetic and toxicological properties. Clinical trials are underway for both solid and haematologic malignancies using anti-CD47 antibodies and recombinant SIRPα proteins. Since the CD47/SIRPα axis also limits the efficacy of tumour-opsonising antibodies, additional trials will examine their potential synergy with agents such as rituximab, cetuximab and trastuzumab. Phagocytosis in response to CD47/SIRPα-blocking agents results in antigen uptake and presentation, thereby linking the innate and adaptive immune systems. CD47/SIRPα blocking therapies may therefore synergise with immune checkpoint inhibitors that target the adaptive immune system. As a critical regulator of macrophage phagocytosis and activation, the potential applications of CD47/SIRPα blocking therapies extend beyond human cancer. They may be useful for the treatment of infectious disease, conditioning for stem cell transplant, and many other clinical indications.
Collapse
Affiliation(s)
- Kipp Weiskopf
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
77
|
Nairz M, Theurl I, Swirski FK, Weiss G. "Pumping iron"-how macrophages handle iron at the systemic, microenvironmental, and cellular levels. Pflugers Arch 2017; 469:397-418. [PMID: 28251312 PMCID: PMC5362662 DOI: 10.1007/s00424-017-1944-8] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/25/2017] [Accepted: 01/29/2017] [Indexed: 12/12/2022]
Abstract
Macrophages reside in virtually every organ. First arising during embryogenesis, macrophages replenish themselves in the adult through a combination of self-renewal and influx of bone marrow-derived monocytes. As large phagocytic cells, macrophages participate in innate immunity while contributing to tissue-specific homeostatic functions. Among the key metabolic tasks are senescent red blood cell recycling, free heme detoxification, and provision of iron for de novo hemoglobin synthesis. While this systemic mechanism involves the shuttling of iron between spleen, liver, and bone marrow through the concerted function of defined macrophage populations, similar circuits appear to exist within the microenvironment of other organs. The high turnover of iron is the prerequisite for continuous erythropoiesis and tissue integrity but challenges macrophages’ ability to maintain cellular iron homeostasis and immune function. This review provides a brief overview of systemic, microenvironmental, and cellular aspects of macrophage iron handling with a focus on exciting and unresolved questions in the field.
Collapse
Affiliation(s)
- Manfred Nairz
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Anichstr. 35, 6020, Innsbruck, Austria. .,Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA. .,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Igor Theurl
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Anichstr. 35, 6020, Innsbruck, Austria
| | - Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Guenter Weiss
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Anichstr. 35, 6020, Innsbruck, Austria.
| |
Collapse
|
78
|
Gallagher S, Turman S, Lekstrom K, Wilson S, Herbst R, Wang Y. CD47 limits antibody dependent phagocytosis against non-malignant B cells. Mol Immunol 2017; 85:57-65. [PMID: 28208074 DOI: 10.1016/j.molimm.2017.01.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/24/2017] [Accepted: 01/25/2017] [Indexed: 01/21/2023]
Abstract
Recent studies have demonstrated the importance of CD47 in protecting malignant B cells from antibody dependent cellular phagocytosis (ADCP). Combined treatment of anti-CD47 and -CD20 antibodies synergistically augment elimination of tumor B cells in xenograft mouse models. This has led to the development of novel reagents that can potentially enhance killing of malignant B cells in patients. B cell depleting therapy is also a promising treatment for autoimmune patients. In the current study, we aimed to investigate whether or not CD47 protects non-malignant B cells from ADCP. We show that CD47 is expressed on all B cells in mice, with the highest level on plasma cells in bone marrow and spleen. Although its expression is dispensable for B cell development in mice, CD47 on B cells limits antibody mediated phagocytosis. B cell depletion following in vivo anti-CD19 treatment is more efficient in CD47-/- mice than in wild type mice. In vitro, both naïve and activated B cells from CD47-/- mice are more sensitive to ADCP than wild type B cells. Lastly, we show in an ADCP assay that blocking CD47 can enhance anti-CD19 antibody mediated phagocytosis of wild type B cells. These results suggest that in addition to its already demonstrated benefit in cancer, targeting CD47 may be used as an adjunct in combination with B cell depletion antibodies for treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Sandra Gallagher
- Department of Oncology Research, MedImmune, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Sean Turman
- Department of Oncology Research, MedImmune, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Kristen Lekstrom
- Department of Protein Science, MedImmune, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Susan Wilson
- Department of Protein Science, MedImmune, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Ronald Herbst
- Department of Oncology Research, MedImmune, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Yue Wang
- Department of Oncology Research, MedImmune, One MedImmune Way, Gaithersburg, MD 20878, USA.
| |
Collapse
|
79
|
Klei TRL, Meinderts SM, van den Berg TK, van Bruggen R. From the Cradle to the Grave: The Role of Macrophages in Erythropoiesis and Erythrophagocytosis. Front Immunol 2017; 8:73. [PMID: 28210260 PMCID: PMC5288342 DOI: 10.3389/fimmu.2017.00073] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/17/2017] [Indexed: 12/13/2022] Open
Abstract
Erythropoiesis is a highly regulated process where sequential events ensure the proper differentiation of hematopoietic stem cells into, ultimately, red blood cells (RBCs). Macrophages in the bone marrow play an important role in hematopoiesis by providing signals that induce differentiation and proliferation of the earliest committed erythroid progenitors. Subsequent differentiation toward the erythroblast stage is accompanied by the formation of so-called erythroblastic islands where a central macrophage provides further cues to induce erythroblast differentiation, expansion, and hemoglobinization. Finally, erythroblasts extrude their nuclei that are phagocytosed by macrophages whereas the reticulocytes are released into the circulation. While in circulation, RBCs slowly accumulate damage that is repaired by macrophages of the spleen. Finally, after 120 days of circulation, senescent RBCs are removed from the circulation by splenic and liver macrophages. Macrophages are thus important for RBCs throughout their lifespan. Finally, in a range of diseases, the delicate interplay between macrophages and both developing and mature RBCs is disturbed. Here, we review the current knowledge on the contribution of macrophages to erythropoiesis and erythrophagocytosis in health and disease.
Collapse
Affiliation(s)
- Thomas R L Klei
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, University of Amsterdam , Amsterdam , Netherlands
| | - Sanne M Meinderts
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, University of Amsterdam , Amsterdam , Netherlands
| | - Timo K van den Berg
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, University of Amsterdam , Amsterdam , Netherlands
| | - Robin van Bruggen
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, University of Amsterdam , Amsterdam , Netherlands
| |
Collapse
|
80
|
Liu J, Santhanakrishnan M, Natarajan P, Gibb DR, Eisenbarth SC, Tormey CA, Siddon AJ, Stowell SR, Branch DR, Hendrickson JE. Antigen modulation as a potential mechanism of anti-KEL immunoprophylaxis in mice. Blood 2016; 128:3159-3168. [PMID: 27688803 PMCID: PMC5201095 DOI: 10.1182/blood-2016-06-724732] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/26/2016] [Indexed: 12/14/2022] Open
Abstract
Red blood cell (RBC) alloimmunization is a serious complication of transfusion or pregnancy. Despite the widespread use of Rh immune globulin to prevent pregnancy associated anti-D alloimmunization, its mechanism of action remains elusive. We have previously described a murine model in which immunoprophylaxis with polyclonal anti-KEL sera prevents alloimmunization in wild-type recipients transfused with transgenic murine RBCs expressing the human KEL glycoprotein. To investigate the mechanism of action, we have now evaluated the outcome of immunoprophylaxis treatment in mice lacking Fcγ receptors (FcγRs), complement (C3), both, or none. Whereas polyclonal anti-KEL sera completely prevented alloimmunization in wild-type and single-knockout (KO) mice lacking FcγRs or C3, double-KO mice lacking both FcγRs and C3 became alloimmunized despite immunoprophylaxis. Rapid clearance of essentially all transfused RBCs with detectable KEL glycoprotein antigen occurred within 24 hours in wild-type and single-KO recipients treated with immunoprophylaxis, with the transfused RBCs remaining in circulation having minimal KEL glycoprotein antigen detectable by flow cytometry or western blot. In contrast, transfused RBCs with the KEL glycoprotein antigen fully intact continued to circulate for days in double-KO mice despite treatment with immunoprophylaxis. Further, in vitro phagocytosis assays showed no consumption of opsonized murine RBCs by double-KO splenocytes. Taken in combination, our data suggest that modulation of the KEL antigen (and potentially RBC clearance) by redundant recipient pathways involving both FcγRs and C3 may be critical to the mechanism of action of polyclonal anti-KEL immunoprophylaxis. These findings could have implications for the development of immunoprophylaxis programs in humans.
Collapse
Affiliation(s)
| | | | | | | | - Stephanie C Eisenbarth
- Department of Laboratory Medicine and
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Christopher A Tormey
- Department of Laboratory Medicine and
- Pathology and Laboratory Medicine Service, VA Connecticut Healthcare System, West Haven, CT
| | - Alexa J Siddon
- Department of Laboratory Medicine and
- Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Sean R Stowell
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA
| | - Donald R Branch
- Centre for Innovation, Canadian Blood Services, Toronto, ON, Canada; and
| | - Jeanne E Hendrickson
- Department of Laboratory Medicine and
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
81
|
Abstract
ABSTRACT
The aim of this review is to provide a coherent framework for understanding dendritic cells (DCs). It has seven sections. The introduction provides an overview of the immune system and essential concepts, particularly for the nonspecialist reader. Next, the “History” section outlines the early evolution of ideas about DCs and highlights some sources of confusion that still exist today. The “Lineages” section then focuses on five different populations of DCs: two subsets of “classical” DCs, plasmacytoid DCs, monocyte-derived DCs, and Langerhans cells. It highlights some cellular and molecular specializations of each, and also notes other DC subsets that have been proposed. The following “Tissues” section discusses the distribution and behavior of different DC subsets within nonlymphoid and secondary lymphoid tissues that are connected by DC migration pathways between them. In the “Tolerance” section, the role of DCs in central and peripheral tolerance is considered, including their ability to drive the differentiation of different populations of regulatory T cells. In contrast, the “Immunity” section considers the roles of DCs in sensing of infection and tissue damage, the initiation of primary responses, the T-cell effector phase, and the induction of immunological memory. The concluding section provides some speculative ideas about the evolution of DCs. It also revisits earlier concepts of generation of diversity and clonal selection in terms of DCs driving the evolution of T-cell responses. Throughout, this review highlights certain areas of uncertainty and suggests some avenues for future investigation.
Collapse
|
82
|
Bian Z, Shi L, Guo YL, Lv Z, Tang C, Niu S, Tremblay A, Venkataramani M, Culpepper C, Li L, Zhou Z, Mansour A, Zhang Y, Gewirtz A, Kidder K, Zen K, Liu Y. Cd47-Sirpα interaction and IL-10 constrain inflammation-induced macrophage phagocytosis of healthy self-cells. Proc Natl Acad Sci U S A 2016; 113:E5434-43. [PMID: 27578867 PMCID: PMC5027463 DOI: 10.1073/pnas.1521069113] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Rapid clearance of adoptively transferred Cd47-null (Cd47(-/-)) cells in congeneic WT mice suggests a critical self-recognition mechanism, in which CD47 is the ubiquitous marker of self, and its interaction with macrophage signal regulatory protein α (SIRPα) triggers inhibitory signaling through SIRPα cytoplasmic immunoreceptor tyrosine-based inhibition motifs and tyrosine phosphatase SHP-1/2. However, instead of displaying self-destruction phenotypes, Cd47(-/-) mice manifest no, or only mild, macrophage phagocytosis toward self-cells except under the nonobese diabetic background. Studying our recently established Sirpα-KO (Sirpα(-/-)) mice, as well as Cd47(-/-) mice, we reveal additional activation and inhibitory mechanisms besides the CD47-SIRPα axis dominantly controlling macrophage behavior. Sirpα(-/-) mice and Cd47(-/-) mice, although being normally healthy, develop severe anemia and splenomegaly under chronic colitis, peritonitis, cytokine treatments, and CFA-/LPS-induced inflammation, owing to splenic macrophages phagocytizing self-red blood cells. Ex vivo phagocytosis assays confirmed general inactivity of macrophages from Sirpα(-/-) or Cd47(-/-) mice toward healthy self-cells, whereas they aggressively attack toward bacteria, zymosan, apoptotic, and immune complex-bound cells; however, treating these macrophages with IL-17, LPS, IL-6, IL-1β, and TNFα, but not IFNγ, dramatically initiates potent phagocytosis toward self-cells, for which only the Cd47-Sirpα interaction restrains. Even for macrophages from WT mice, phagocytosis toward Cd47(-/-) cells does not occur without phagocytic activation. Mechanistic studies suggest a PKC-Syk-mediated signaling pathway, to which IL-10 conversely inhibits, is required for activating macrophage self-targeting, followed by phagocytosis independent of calreticulin Moreover, we identified spleen red pulp to be one specific tissue that provides stimuli constantly activating macrophage phagocytosis albeit lacking in Cd47(-/-) or Sirpα(-/-) mice.
Collapse
Affiliation(s)
- Zhen Bian
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Advanced Institute for Life Sciences, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Lei Shi
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302
| | - Ya-Lan Guo
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302
| | - Zhiyuan Lv
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302
| | - Cong Tang
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302
| | - Shuo Niu
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302
| | - Alexandra Tremblay
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302
| | - Mahathi Venkataramani
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302
| | - Courtney Culpepper
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302
| | - Limin Li
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Advanced Institute for Life Sciences, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Zhen Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Advanced Institute for Life Sciences, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Ahmed Mansour
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, Life Science Institute (LSI) Immunology Programme, National University of Singapore, Singapore 117456
| | - Andrew Gewirtz
- Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA 30303
| | - Koby Kidder
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302; Department of Cell Biology, Rutgers University, New Brunswick, NJ 08901
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Advanced Institute for Life Sciences, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Yuan Liu
- Program of Immunology and Cell Biology, Department of Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA 30302; Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA 30303;
| |
Collapse
|
83
|
Gibb DR, Calabro S, Liu D, Tormey CA, Spitalnik SL, Zimring JC, Hendrickson JE, Hod EA, Eisenbarth SC. The Nlrp3 Inflammasome Does Not Regulate Alloimmunization to Transfused Red Blood Cells in Mice. EBioMedicine 2016; 9:77-86. [PMID: 27345021 PMCID: PMC4972549 DOI: 10.1016/j.ebiom.2016.06.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/02/2016] [Accepted: 06/05/2016] [Indexed: 02/07/2023] Open
Abstract
Red blood cell (RBC) transfusions are essential for patients with hematological disorders and bone marrow failure syndromes. Despite ABO matching, RBC transfusions can lead to production of alloantibodies against “minor” blood group antigens. Non-ABO alloimmunization is a leading cause of transfusion-associated mortality in the U.S. Despite its clinical importance, little is known about the immunological factors that promote alloimmunization. Prior studies indicate that inflammatory conditions place patients at higher risk for alloimmunization. Additionally, co-exposure to pro-inflammatory pathogen associated molecular patterns (PAMPs) promotes alloimmunization in animal models, suggesting that RBC alloimmunization depends on innate immune cell activation. However, the specific innate immune stimuli and sensors that induce a T cell-dependent alloantibody response to transfused RBCs have not been identified. The NLRP3 inflammasome senses chemically diverse PAMPs and damage associated molecular patterns (DAMPs), including extracellular ATP and iron-containing heme. We hypothesized that activation of the NLRP3 inflammasome by endogenous DAMPs from RBCs promotes the alloimmune response to a sterile RBC transfusion. Using genetically modified mice lacking either NLRP3 or multiple downstream inflammasome response elements, we ruled out a role for the NLRP3 inflammasome or any Caspase-1 or -11 dependent inflammasome in regulating RBC alloantibody production to a model antigen. Transfusion of stored red blood cells (RBCs) induces proinflammatory cytokine production and alloimmunization to an RBC antigen in mice. Transfusion of stored RBCs, regardless of alloantigen expression, activates conventional dendritic cells in the spleen. NOD-like receptor (NLR) inflammasomes, including NLRP3, do not regulate inflammation and alloimmunization induced by stored RBCs.
Following a blood transfusion, the immune system may produce antibodies that have detrimental effects. To understand how the immune system recognizes factors in transfused blood, we examined the immune response of mice lacking important inflammatory molecules, called inflammasomes. The results demonstrate that inflammasomes do not affect the production of potentially harmful antibodies that recognize transfused red blood cells.
Collapse
Affiliation(s)
- David R Gibb
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Samuele Calabro
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Dong Liu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Christopher A Tormey
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Steven L Spitalnik
- Department of Pathology & Cell Biology, Columbia University Medical Center-New York Presbyterian Hospital, NY, New York 10032, USA
| | - James C Zimring
- Bloodworks NW Research Institute, Seattle, WA 98102, USA; Department of Laboratory Medicine, University of Washington School of Medicine, Seattle, WA 98102, USA; Department of Internal Medicine-Division of Hematology, University of Washington School of Medicine, Seattle, WA 98102, USA
| | - Jeanne E Hendrickson
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Eldad A Hod
- Department of Pathology & Cell Biology, Columbia University Medical Center-New York Presbyterian Hospital, NY, New York 10032, USA.
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
84
|
Calabro S, Gallman A, Gowthaman U, Liu D, Chen P, Liu J, Krishnaswamy JK, Nascimento MSL, Xu L, Patel SR, Williams A, Tormey CA, Hod EA, Spitalnik SL, Zimring JC, Hendrickson JE, Stowell SR, Eisenbarth SC. Bridging channel dendritic cells induce immunity to transfused red blood cells. J Exp Med 2016; 213:887-96. [PMID: 27185856 PMCID: PMC4886363 DOI: 10.1084/jem.20151720] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 04/06/2016] [Indexed: 11/30/2022] Open
Abstract
Calabro et al. show that 33D1+ dendritic cells present in the bridging channel of the spleen are essential for alloantibody response to transfused red blood cells. Red blood cell (RBC) transfusion is a life-saving therapeutic tool. However, a major complication in transfusion recipients is the generation of antibodies against non-ABO alloantigens on donor RBCs, potentially resulting in hemolysis and renal failure. Long-lived antibody responses typically require CD4+ T cell help and, in murine transfusion models, alloimmunization requires a spleen. Yet, it is not known how RBC-derived antigens are presented to naive T cells in the spleen. We sought to answer whether splenic dendritic cells (DCs) were essential for T cell priming to RBC alloantigens. Transient deletion of conventional DCs at the time of transfusion or splenic DC preactivation before RBC transfusion abrogated T and B cell responses to allogeneic RBCs, even though transfused RBCs persisted in the circulation for weeks. Although all splenic DCs phagocytosed RBCs and activated RBC-specific CD4+ T cells in vitro, only bridging channel 33D1+ DCs were required for alloimmunization in vivo. In contrast, deletion of XCR1+CD8+ DCs did not alter the immune response to RBCs. Our work suggests that blocking the function of one DC subset during a narrow window of time during RBC transfusion could potentially prevent the detrimental immune response that occurs in patients who require lifelong RBC transfusion support.
Collapse
Affiliation(s)
- Samuele Calabro
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520 Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Antonia Gallman
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520 Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Uthaman Gowthaman
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520 Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Dong Liu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520 Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Pei Chen
- Department of Neurology, The First Affiliated Hospital of Sun Yat-Sen University, Yuexiu, Guangzhou, Guangdong, 510080, China
| | - Jingchun Liu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Jayendra Kumar Krishnaswamy
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520 Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Manuela Sales L Nascimento
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520 Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520 Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Lan Xu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520 Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Seema R Patel
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322
| | - Adam Williams
- The Jackson Laboratory for Genomic Medicine, Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06030
| | - Christopher A Tormey
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Eldad A Hod
- Department of Pathology and Cell Biology, Columbia University Medical Center-New York Presbyterian Hospital, New York, NY 10032
| | - Steven L Spitalnik
- Department of Pathology and Cell Biology, Columbia University Medical Center-New York Presbyterian Hospital, New York, NY 10032
| | - James C Zimring
- Bloodworks NW Research Institute, University of Washington School of Medicine, Seattle, WA 98102 Department of Laboratory Medicine, University of Washington School of Medicine, Seattle, WA 98102 Division of Hematology, Department of Internal Medicine, University of Washington School of Medicine, Seattle, WA 98102
| | - Jeanne E Hendrickson
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520 Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520
| | - Sean R Stowell
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520 Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| |
Collapse
|
85
|
Li J, Lu E, Yi T, Cyster JG. EBI2 augments Tfh cell fate by promoting interaction with IL-2-quenching dendritic cells. Nature 2016; 533:110-4. [PMID: 27147029 PMCID: PMC4883664 DOI: 10.1038/nature17947] [Citation(s) in RCA: 218] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/29/2016] [Indexed: 01/08/2023]
Abstract
T follicular helper (Tfh) cells are a subset of T cells carrying the CD4 antigen; they are important in supporting plasma cell and germinal centre responses. The initial induction of Tfh cell properties occurs within the first few days after activation by antigen recognition on dendritic cells, although how dendritic cells promote this cell-fate decision is not fully understood. Moreover, although Tfh cells are uniquely defined by expression of the follicle-homing receptor CXCR5 (refs 1, 2), the guidance receptor promoting the earlier localization of activated T cells at the interface of the B-cell follicle and T zone has been unclear. Here we show that the G-protein-coupled receptor EBI2 (GPR183) and its ligand 7α,25-dihydroxycholesterol mediate positioning of activated CD4 T cells at the interface of the follicle and T zone. In this location they interact with activated dendritic cells and are exposed to Tfh-cell-promoting inducible co-stimulator (ICOS) ligand. Interleukin-2 (IL-2) is a cytokine that has multiple influences on T-cell fate, including negative regulation of Tfh cell differentiation. We demonstrate that activated dendritic cells in the outer T zone further augment Tfh cell differentiation by producing membrane and soluble forms of CD25, the IL-2 receptor α-chain, and quenching T-cell-derived IL-2. Mice lacking EBI2 in T cells or CD25 in dendritic cells have reduced Tfh cells and mount defective T-cell-dependent plasma cell and germinal centre responses. These findings demonstrate that distinct niches within the lymphoid organ T zone support distinct cell fate decisions, and they establish a function for dendritic-cell-derived CD25 in controlling IL-2 availability and T-cell differentiation.
Collapse
Affiliation(s)
- Jianhua Li
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California 94143, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, California 94143, USA
- Key Laboratory of Medical Molecular Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Erick Lu
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California 94143, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, California 94143, USA
| | - Tangsheng Yi
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California 94143, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, California 94143, USA
| | - Jason G Cyster
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California 94143, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
86
|
Larsson A, Hult A, Nilsson A, Olsson M, Oldenborg PA. Red blood cells with elevated cytoplasmic Ca2+are primarily taken up by splenic marginal zone macrophages and CD207+ dendritic cells. Transfusion 2016; 56:1834-44. [DOI: 10.1111/trf.13612] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 02/23/2016] [Accepted: 03/03/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Anders Larsson
- Department of Integrative Medical Biology and the; Umeå University; Umeå Sweden
| | - Andreas Hult
- Department of Community Medicine and Rehabilitation, Section for Sports Medicine; Umeå University; Umeå Sweden
| | - Anna Nilsson
- Department of Integrative Medical Biology and the; Umeå University; Umeå Sweden
| | - Mattias Olsson
- Department of Integrative Medical Biology and the; Umeå University; Umeå Sweden
| | - Per-Arne Oldenborg
- Department of Integrative Medical Biology and the; Umeå University; Umeå Sweden
| |
Collapse
|
87
|
Durable antitumor responses to CD47 blockade require adaptive immune stimulation. Proc Natl Acad Sci U S A 2016; 113:E2646-54. [PMID: 27091975 DOI: 10.1073/pnas.1604268113] [Citation(s) in RCA: 273] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Therapeutic antitumor antibodies treat cancer by mobilizing both innate and adaptive immunity. CD47 is an antiphagocytic ligand exploited by tumor cells to blunt antibody effector functions by transmitting an inhibitory signal through its receptor signal regulatory protein alpha (SIRPα). Interference with the CD47-SIRPα interaction synergizes with tumor-specific monoclonal antibodies to eliminate human tumor xenografts by enhancing macrophage-mediated antibody-dependent cellular phagocytosis (ADCP), but synergy between CD47 blockade and ADCP has yet to be demonstrated in immunocompetent hosts. Here, we show that CD47 blockade alone or in combination with a tumor-specific antibody fails to generate antitumor immunity against syngeneic B16F10 tumors in mice. Durable tumor immunity required programmed death-ligand 1 (PD-L1) blockade in combination with an antitumor antibody, with incorporation of CD47 antagonism substantially improving response rates. Our results highlight an underappreciated contribution of the adaptive immune system to anti-CD47 adjuvant therapy and suggest that targeting both innate and adaptive immune checkpoints can potentiate the vaccinal effect of antitumor antibody therapy.
Collapse
|
88
|
|