1
|
Deng WQ, Ye ZH, Tang Z, Zhang XL, Lu JJ. Beyond cancer: The potential application of CD47-based therapy in non-cancer diseases. Acta Pharm Sin B 2025; 15:757-791. [PMID: 40177549 PMCID: PMC11959971 DOI: 10.1016/j.apsb.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/10/2024] [Accepted: 11/22/2024] [Indexed: 04/05/2025] Open
Abstract
CD47 is an immune checkpoint widely regarded as a 'don't eat me' signal. CD47-based anti-cancer therapy has received considerable attention, with a significant number of clinical trials conducted. While anti-cancer therapies based on CD47 remain a focal point of interest among researchers, it is noteworthy that an increasing number of studies have found that CD47-based therapy ameliorated the pathological status of non-cancer diseases. This review aims to provide an overview of the recent progress in comprehending the role of CD47-based therapy in non-cancer diseases, including diseases of the circulatory system, nervous system, digestive system, and so on. Furthermore, we sought to delineate the promising mechanisms of CD47-based therapy in treating non-cancer diseases. Our findings suggest that CD47-based agents may exert their effect by regulating phagocytosis, regulating T cells, dendritic cells, and neutrophils, and regulating the secretion of cytokines and chemokines. Additionally, we put forward the orientation of further research to bring to light the potential of CD47 and its binding partners as a target in non-cancer diseases.
Collapse
Affiliation(s)
- Wei-Qing Deng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Zi-Han Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Zhenghai Tang
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
| | - Xiao-Lei Zhang
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao 999078, China
| |
Collapse
|
2
|
Koskinen Holm C, Rosendahl S, Oldenborg PA, Lundberg P. The expression of signal regulatory protein alpha (SIRPα) in periodontal cells and tissue. Acta Odontol Scand 2024; 83:486-492. [PMID: 39258954 PMCID: PMC11409820 DOI: 10.2340/aos.v83.41391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/29/2024] [Indexed: 09/12/2024]
Abstract
Signal regulatory protein alpha (SIRPα) is mainly expressed by cells of myeloid origin. This membrane glycoprotein is shown to be involved in regulation of different inflammatory conditions, such as colitis and arthritis. However, SIRPα has not been investigated in relationship to periodontitis, an inflammatory condition affecting the tooth supporting tissues. We aim to investigate if resident cells in the periodontium express SIRPα and whether a possible expression is affected by inflammatory conditions. Primary human keratinocytes, fibroblasts, periodontal ligament cells, and osteoblasts were cultured with or without the pro-inflammatory cytokines tumor necrosis factor alpha (TNF-α) or interleukin-1-beta (IL-1β). All different periodontal cell types showed a basal mRNA expression of SIRPα. Pro-inflammatory cytokines induced a 2-3-fold significant increase in SIRPα expression in both cultured human gingival fibroblasts and osteoblasts but neither in keratinocytes nor in periodontal ligament cells. Tissue sections from human gingival tissue biopsies were histochemically stained for SIRPα. Epithelial keratinocytes and gingival fibroblasts stained positive in sections from periodontally healthy as well as in sections from periodontitis. In periodontitis sections, infiltrating leukocytes stained positive for SIRPα. We highlight our finding that oral keratinocytes, gingival fibroblasts, and periodontal ligament cells do express SIRPα, as this has not been presented before. The fact that inflammatory stimulation of gingival fibroblasts increased the expression of SIRPα, while an increased expression by gingival fibroblasts in periodontitis tissue in situ could not be detected, is indeed contradictory.
Collapse
Affiliation(s)
- Cecilia Koskinen Holm
- Department of Odontology, Section of Molecular Periodontology, Umeå University, Umeå, Sweden; Department of Odontology, Section of Oral and Maxillofacial Surgery, Umeå University, Umeå, Sweden.
| | - Sara Rosendahl
- Department of Odontology, Section of Molecular Periodontology, Umeå University, Umeå, Sweden
| | - Per-Arne Oldenborg
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Pernilla Lundberg
- Department of Odontology, Section of Molecular Periodontology, Umeå University, Umeå, Sweden
| |
Collapse
|
3
|
Huang F, Su Z, Huang Y, Huang Y, Zhou C, Feng S, Qin X, Xie X, Liu C, Yu C. Exploration of the combined role of immune checkpoints and immune cells in the diagnosis and treatment of ankylosing spondylitis: a preliminary study immune checkpoints in ankylosing spondylitis. Arthritis Res Ther 2024; 26:115. [PMID: 38835033 DOI: 10.1186/s13075-024-03341-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/12/2024] [Indexed: 06/06/2024] Open
Abstract
OBJECTIVE Immune checkpoints have emerged as promising therapeutic targets for autoimmune diseases. However, the specific roles of immune checkpoints in the pathophysiology of ankylosing spondylitis (AS) remain unclear. METHODS Hip ligament samples were obtained from two patient groups: those with AS and femoral head deformity, and those with femoral head necrosis but without AS, undergoing hip arthroplasty. Label-Free Quantification (LFQ) Protein Park Analysis was used to identify the protein composition of the ligaments. Peripheral blood samples of 104 AS patients from public database were used to validate the expression of key proteins. KEGG, GO, and GSVA were employed to explore potential pathways regulated by immune checkpoints in AS progression. xCell was used to calculate cell infiltration levels, LASSO regression was applied to select key cells, and the correlation between immune checkpoints and immune cells was analyzed. Drug sensitivity analysis was conducted to identify potential therapeutic drugs targeting immune checkpoints in AS. The expression of key genes was validated through immunohistochemistry (IHC). RESULTS HLA-DMB and HLA-DPA1 were downregulated in the ligaments of AS and this has been validated through peripheral blood datasets and IHC. Significant differences in expression were observed in CD8 + Tcm, CD8 + T cells, CD8 + Tem, osteoblasts, Th1 cells, and CD8 + naive T cells in AS. The infiltration levels of CD8 + Tcm and CD8 + naive T cells were significantly positively correlated with the expression levels of HLA-DMB and HLA-DPA1. Immune cell selection using LASSO regression showed good predictive ability for AS, with AUC values of 0.98, 0.81, and 0.75 for the three prediction models, respectively. Furthermore, this study found that HLA-DMB and HLA-DPA1 are involved in Th17 cell differentiation, and both Th17 cell differentiation and the NF-kappa B signaling pathway are activated in the AS group. Drug sensitivity analysis showed that AS patients are more sensitive to drugs such as doramapimod and GSK269962A. CONCLUSION Immune checkpoints and immune cells could serve as avenues for exploring diagnostic and therapeutic strategies for AS.
Collapse
Affiliation(s)
- Feihong Huang
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Zhiping Su
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Yibin Huang
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Yuxiang Huang
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Chengyu Zhou
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Sitan Feng
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Xiong Qin
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Xi Xie
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Chong Liu
- Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China.
| | - Chaojie Yu
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, China.
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China.
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China.
| |
Collapse
|
4
|
Ye X, Li H, Anjum K, Zhong X, Miao S, Zheng G, Liu W, Li L. Dual Role of Indoles Derived From Intestinal Microbiota on Human Health. Front Immunol 2022; 13:903526. [PMID: 35784338 PMCID: PMC9248744 DOI: 10.3389/fimmu.2022.903526] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/23/2022] [Indexed: 12/12/2022] Open
Abstract
Endogenous indole and its derivatives (indoles), considered as promising N-substituted heterocyclic compounds, are tryptophan metabolites derived from intestinal microbiota and exhibit a range of biological activities. Recent studies indicate that indoles contribute to maintaining the biological barrier of the human intestine, which exert the anti-inflammatory activities mainly through activating AhR and PXR receptors to affect the immune system’s function, significantly improving intestinal health (inflammatory bowel disease, hemorrhagic colitis, colorectal cancer) and further promote human health (diabetes mellitus, central system inflammation, and vascular regulation). However, the revealed toxic influences cannot be ignored. Indoxyl sulfate, an indole derivative, performs nephrotoxicity and cardiovascular toxicity. We addressed the interaction between indoles and intestinal microbiota and the indoles’ effects on human health as double-edged swords. This review provides scientific bases for the correlation of indoles with diseases moreover highlights several directions for subsequent indoles-related studies.
Collapse
Affiliation(s)
- Xuewei Ye
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
- Department of Basic Medical Sciences, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Haiyi Li
- Department of Basic Medical Sciences, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Komal Anjum
- Department of Medicine and pharmacy, Ocean University of China, Qingdao, China
| | - Xinye Zhong
- Department of Basic Medical Sciences, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Shuping Miao
- Department of Basic Medical Sciences, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Guowan Zheng
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Wei Liu
- Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
- *Correspondence: Lanjuan Li, ; Wei Liu,
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
- *Correspondence: Lanjuan Li, ; Wei Liu,
| |
Collapse
|
5
|
Shen Q, Zhao L, Pan L, Li D, Chen G, Chen Z, Jiang Z. Soluble SIRP-Alpha Promotes Murine Acute Lung Injury Through Suppressing Macrophage Phagocytosis. Front Immunol 2022; 13:865579. [PMID: 35634325 PMCID: PMC9133620 DOI: 10.3389/fimmu.2022.865579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/13/2022] [Indexed: 11/17/2022] Open
Abstract
Soluble signal regulatory protein-alpha (SIRP-alpha) is elevated in bronchoalveolar lavage (BAL) of mice with lipopolysaccharides (LPS)-induced acute lung injury (ALI). To define the role of soluble SIRP-alpha in the pathogenesis of ALI, we established murine ALI in wild-type (WT) and SIRP-alpha knock-out (KO) mice by intratracheal administration of LPS. The results indicated that lack of SIRP-alpha significantly reduced the pathogenesis of ALI, in association with attenuated lung inflammation, infiltration of neutrophils and expression of pro-inflammatory cytokines in mice. In addition, lack of SIRP-alpha reduced the expression of pro-inflammatory cytokines in LPS-treated bone marrow-derived macrophages (BMDMs) from KO mice, accompanied with improved macrophage phagocytosis. Blockade of soluble SIRP-alpha activity in ALI BAL by anti-SIRP-alpha antibody (aSIRP) effectively reduced the expression of TNF-alpha and IL-6 mRNA transcripts and proteins, improved macrophage phagocytosis in vitro. In addition, lack of SIRP-alpha reduced activation of Src homology 2 domain-containing protein tyrosine phosphatase 1 (SHP-1) and improved activation of signal transducer and activator of transcription-3 (STAT3) and STAT6. Suppression of SHP-1 activity by tyrosine phosphatase inhibitor 1 (TPI-1) increased activation of STAT3 and STAT6, and improved macrophage phagocytosis, that was effectively reversed by STAT3 and STAT6 inhibitors. Thereby, SIRP-alpha suppressed macrophage phagocytosis through activation of SHP-1, subsequently inhibiting downstream STAT3 and STAT6 signaling. Lack of SIRP-alpha attenuated murine ALI possibly through increasing phagocytosis, and improving STAT3 and STAT6 signaling in macrophages. SIRP-alpha would be promising biomarker and molecular target in the treatment of murine ALI and patients with acute respiratory distress syndrome (ARDS).
Collapse
Affiliation(s)
- Qinjun Shen
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Li Zhao
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Linyue Pan
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dandan Li
- Department of Pulmonary and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Gang Chen
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhihong Chen
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhilong Jiang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Jarr KU, Ye J, Kojima Y, Ye Z, Gao H, Schmid S, Luo L, Baylis RA, Lotfi M, Lopez N, Eberhard AV, Smith BR, Weissman IL, Maegdefessel L, Leeper NJ. The pleiotropic benefits of statins include the ability to reduce CD47 and amplify the effect of pro-efferocytic therapies in atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2022; 1:253-262. [PMID: 35990913 PMCID: PMC9390974 DOI: 10.1038/s44161-022-00023-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 01/26/2022] [Indexed: 01/20/2023]
Abstract
The pleiotropic benefits of statins may result from their impact on vascular inflammation. The molecular process underlying this phenomenon is not fully elucidated. Here, RNA sequencing designed to investigate gene expression patterns following CD47-SIRPα inhibition identifies a link between statins, efferocytosis, and vascular inflammation. In vivo and in vitro studies provide evidence that statins augment programmed cell removal by inhibiting the nuclear translocation of NFκB1 p50 and suppressing the expression of the critical 'don't eat me' molecule, CD47. Statins amplify the phagocytic capacity of macrophages, and thus the anti-atherosclerotic effects of CD47-SIRPα blockade, in an additive manner. Analyses of clinical biobank specimens suggest a similar link between statins and CD47 expression in humans, highlighting the potential translational implications. Taken together, our findings identify efferocytosis and CD47 as pivotal mediators of statin pleiotropy. In turn, statins amplify the anti-atherosclerotic effects of pro-phagocytic therapies independently of any lipid-lowering effect.
Collapse
Affiliation(s)
- Kai-Uwe Jarr
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Jianqin Ye
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Yoko Kojima
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Zhongde Ye
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Hua Gao
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Sofie Schmid
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Lingfeng Luo
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Richard A. Baylis
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Mozhgan Lotfi
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Nicolas Lopez
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Anne V. Eberhard
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Bryan Ronain Smith
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, United States of America
- Institute for Quantitative Health Science and Engineering, East Lansing, Michigan, United States of America
| | - Irving L. Weissman
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, United States of America
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
- German Center for Cardiovascular Research, Berlin, Germany (DZHK partner site Munich Heart Alliance)
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Nicholas J. Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Stanford Cardiovascular Institute, Stanford University, Stanford, California, United States of America
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
7
|
Nikolakis D, de Voogd FAE, Pruijt MJ, Grootjans J, van de Sande MG, D’Haens GR. The Role of the Lymphatic System in the Pathogenesis and Treatment of Inflammatory Bowel Disease. Int J Mol Sci 2022; 23:ijms23031854. [PMID: 35163775 PMCID: PMC8836364 DOI: 10.3390/ijms23031854] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/26/2022] [Accepted: 02/01/2022] [Indexed: 02/04/2023] Open
Abstract
Although the number of therapeutic options for the treatment of inflammatory bowel disease (IBD) has increased in recent years, patients suffer from decreased quality of life due to non-response or loss of response to the currently available treatments. An increased understanding of the disease’s etiology could provide novel insights for treatment strategies in IBD. Lymphatic system components are generally linked to immune responses and presumably related to inflammatory diseases pathophysiology. This review aims to summarize findings on immune-mediated mechanisms in lymphoid tissues linked with IBD pathogenesis and (potential) novel treatments. Enhanced innate and adaptive immune responses were observed in mesenteric lymph nodes (MLNs) and other lymphoid structures, such as Peyer’s patches, in patients with IBD and in animal models. Furthermore, the phenomenon of lymphatic obstruction in the form of granulomas in MLNs and lymphatic vessels correlates with disease activity. There is also evidence that abnormalities in the lymphatic stromal components and lymph node microbiome are common in IBD and could be exploited therapeutically. Finally, novel agents targeting lymphocyte trafficking have been added to the treatment armamentarium in the field of IBD. Overall, gut-associated lymphoid tissue plays a key role in IBD immunopathogenesis, which could offer novel therapeutic targets.
Collapse
Affiliation(s)
- Dimitrios Nikolakis
- Department of Gastroenterology, Amsterdam Institute for Gastroenterology Endocrinology and Metabolism, Academic Medical Center, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (D.N.); (F.A.E.d.V.); (M.J.P.); (J.G.)
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
- Department of Experimental Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Onassis Foundation, 4 Aeschinou Street, 10558 Athens, Greece
| | - Floris A. E. de Voogd
- Department of Gastroenterology, Amsterdam Institute for Gastroenterology Endocrinology and Metabolism, Academic Medical Center, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (D.N.); (F.A.E.d.V.); (M.J.P.); (J.G.)
| | - Maarten J. Pruijt
- Department of Gastroenterology, Amsterdam Institute for Gastroenterology Endocrinology and Metabolism, Academic Medical Center, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (D.N.); (F.A.E.d.V.); (M.J.P.); (J.G.)
| | - Joep Grootjans
- Department of Gastroenterology, Amsterdam Institute for Gastroenterology Endocrinology and Metabolism, Academic Medical Center, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (D.N.); (F.A.E.d.V.); (M.J.P.); (J.G.)
| | - Marleen G. van de Sande
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
- Department of Experimental Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Geert R. D’Haens
- Department of Gastroenterology, Amsterdam Institute for Gastroenterology Endocrinology and Metabolism, Academic Medical Center, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (D.N.); (F.A.E.d.V.); (M.J.P.); (J.G.)
- Correspondence:
| |
Collapse
|
8
|
Lyu N, Yi JZ, Zhao M. Immunotherapy in older patients with hepatocellular carcinoma. Eur J Cancer 2021; 162:76-98. [PMID: 34954439 DOI: 10.1016/j.ejca.2021.11.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/31/2021] [Accepted: 11/21/2021] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common types of cancer globally and is currently the third leading cause of cancer-related deaths. Recently, immunotherapy using immune checkpoint inhibitors (ICIs) has been shown with encouraging anticancer activity and safety in clinical trials. To reverse the phenomenon of tumours evading immune response, ICIs can be used to stimulate the natural antitumour potential of cancer cells by blocking the relevant checkpoints to activate T cells. However, the components and functions of the immune system may undergo a series of changes with ageing, known as 'immunosenescence,' potentially affecting the antitumour effect and safety of immunotherapy. In the current phase III clinical trials of ICIs including nivolumab, pembrolizumab and atezolizumab, the proportion of patients with HCC older than 65 years in CheckMate 459, KEYNOTE-240 and IMbrave150 is 51%, 58% and 50%, respectively, which is less than 70%-73% of epidemiological investigation. Therefore, the elderly population recruited in clinical trials may not accurately represent the real-world elderly patients with HCC, which affects the extrapolation of the efficacy and safety profile obtained in clinical trials to the elderly population in the real world. This review provides the latest advances in ICIs immuno-treatment available for HCC and relevant information about their therapeutic effects and safety on elderly patients. We discuss the benefits of ICIs for older HCC patients, and relevant recommendations about conducting further clinical trials are proposed for more complete answers to this clinical issue.
Collapse
Affiliation(s)
- Ning Lyu
- Department of Minimally Invasive Interventional Therapy, Liver Cancer Study and Service Group, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jun-Zhe Yi
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ming Zhao
- Department of Minimally Invasive Interventional Therapy, Liver Cancer Study and Service Group, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
9
|
Xu Z, Zeng H, Liu Z, Jin K, Chang Y, Wang Y, Liu L, Zhu Y, Xu L, Wang Z, Guo J, Xu J. Poor clinical outcomes and immunoevasive contexture in SIRPα + tumor-associated macrophages enriched muscle-invasive bladder cancer patients. Urol Oncol 2021; 40:109.e11-109.e20. [PMID: 34600802 DOI: 10.1016/j.urolonc.2021.08.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/31/2021] [Accepted: 08/28/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVES In tumor immune microenvironment, the functions of tumor-associated macrophages (TAMs), including phagocytosis and immunomodulatory, have attracted increasing attention recently. With the discovery of CD47-signal regulatory protein-α (SIRPα) as "don't eat me" signaling pathway, the role of novel subpopulation of TAMs expressing SIRPα has not been fully elucidated in a wide spectrum of solid tumors including bladder cancer. In this study, we investigated the prognostic and predictive implication of SIRPα+ TAMs regarding clinical outcomes and adjuvant chemotherapeutic benefit in muscle-invasive bladder cancer (MIBC), and preliminarily characterized the phenotypic features of SIRPα+ TAMs and its relationship with immune contexture. MATERIALS AND METHODS A total of 141 histochemical MIBC samples from Zhongshan Hospital (ZS), 45 fresh tissue samples, and 391 MIBC patients from TCGA database were enrolled in this study. SIRPα+ TAMs was evaluated by immunohistochemical staining of CD68 and SIRPα, and flow cytometry fluorescence staining. RESULTS Our results illustrated that SIRPα+ TAMs were enriched in MIBC specimens. Patients with high SIRPα+ TAMs infiltration suffered significant poor overall survival and recurrence-free survival (P = 0.0030 and P = 0.0282). SIRPα+ TAMs infiltration was an independent prognosticator in multivariate Cox model. Moreover, adjuvant chemotherapy (ACT) application showed significantly survival benefit in patients with low SIRPα+ TAMs infiltration (P = 0.0135). SIRPα+ TAMs with suppressive phenotype exhibited a positive correlation with immune tolerance and dysfunctional CD8+ T cells in MIBC. CONCLUSIONS SIRPα+ TAMs infiltration indicated poor prognosis and ACT resistance in MIBC. Immunosuppressive SIRPα+ TAMs is closely related to immune evasion with exhausted T cells states, suggesting the prospect of SIRPα+ TAMs as a potential therapeutic target in MIBC.
Collapse
Affiliation(s)
- Ziang Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Han Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhaopei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Kaifeng Jin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yuan Chang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yiwei Wang
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Liu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Le Xu
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zewei Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jiejie Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
10
|
Okunuki Y, Tabor SJ, Lee MY, Connor KM. CD47 Deficiency Ameliorates Ocular Autoimmune Inflammation. Front Immunol 2021; 12:680568. [PMID: 34093583 PMCID: PMC8174453 DOI: 10.3389/fimmu.2021.680568] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/29/2021] [Indexed: 12/20/2022] Open
Abstract
Autoimmune uveitis is a sight-threatening ocular inflammatory condition in which the retina and uveal tissues become a target of autoreactive immune cells. The CD47 is a ubiquitously expressed transmembrane protein which plays multiple roles in fundamental cellular functions including phagocytosis, proliferation, and adhesion. Signal regulatory protein alpha (SIRPα), one of the CD47 ligands, is predominantly expressed in myeloid lineage cells such as dendritic cells (DCs) or macrophages, and CD47-SIRPα signaling pathway is implicated in the development of autoimmune diseases. Our current study demonstrates how CD47 depletion is effective in the prevention of experimental autoimmune uveitis (EAU), an animal model of human autoimmune uveitis, in animals deficient of CD47 (CD47-/- ). Systemic suppression of SIRPα+ DCs in animals deficient in CD47 resulted in the inability of autoreactive CD4+ T cells to develop, which is crucial to induction of EAU. Of interest, retinal microglia, the resident immune cell of the retina, express SIRPα, however these cells were not operative in EAU suppression in response to CD47 depletion. These results identify CD47 as a significant regulator in the development of SIRPα+ DCs that is vital to disease induction in EAU.
Collapse
Affiliation(s)
| | | | | | - Kip M. Connor
- Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
11
|
Christofi M, Le Sommer S, Mölzer C, Klaska IP, Kuffova L, Forrester JV. Low-dose 2-deoxy glucose stabilises tolerogenic dendritic cells and generates potent in vivo immunosuppressive effects. Cell Mol Life Sci 2020; 78:2857-2876. [PMID: 33074350 PMCID: PMC8004500 DOI: 10.1007/s00018-020-03672-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 09/04/2020] [Accepted: 10/05/2020] [Indexed: 01/17/2023]
Abstract
Cell therapies for autoimmune diseases using tolerogenic dendritic cells (tolDC) have been promisingly explored. A major stumbling block has been generating stable tolDC, with low risk of converting to mature immunogenic DC (mDC), exacerbating disease. mDC induction involves a metabolic shift to lactate production from oxidative phosphorylation (OXPHOS) and β-oxidation, the homeostatic energy source for resting DC. Inhibition of glycolysis through the administration of 2-deoxy glucose (2-DG) has been shown to prevent autoimmune disease experimentally but is not clinically feasible. We show here that treatment of mouse bone marrow-derived tolDC ex vivo with low-dose 2-DG (2.5 mM) (2-DGtolDC) induces a stable tolerogenic phenotype demonstrated by their failure to engage lactate production when challenged with mycobacterial antigen (Mtb). ~ 15% of 2-DGtolDC express low levels of MHC class II and 30% express CD86, while they are negative for CD40. 2-DGtolDC also express increased immune checkpoint molecules PDL-1 and SIRP-1α. Antigen-specific T cell proliferation is reduced in response to 2-DGtolDC in vitro. Mtb-stimulated 2-DGtolDC do not engage aerobic glycolysis but respond to challenge via increased OXPHOS. They also have decreased levels of p65 phosphorylation, with increased phosphorylation of the non-canonical p100 pathway. A stable tolDC phenotype is associated with sustained SIRP-1α phosphorylation and p85-AKT and PI3K signalling inhibition. Further, 2-DGtolDC preferentially secrete IL-10 rather than IL-12 upon Mtb-stimulation. Importantly, a single subcutaneous administration of 2-DGtolDC prevented experimental autoimmune uveoretinitis (EAU) in vivo. Inhibiting glycolysis of autologous tolDC prior to transfer may be a useful approach to providing stable tolDC therapy for autoimmune/immune-mediated diseases.
Collapse
Affiliation(s)
- M Christofi
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - S Le Sommer
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - C Mölzer
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK.
| | - I P Klaska
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - L Kuffova
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK.,Eye Clinic, Aberdeen Royal Infirmary, Aberdeen, Scotland, UK
| | - J V Forrester
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK. .,Ocular Immunology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, WA, Australia. .,Centre for Experimental Immunology, Lions Eye Institute, Nedlands, WA, Australia.
| |
Collapse
|
12
|
He Y, Sun X, Rong W, Yang R, Liang H, Qi Y, Li L, Zen K. CD47 is a negative regulator of intestinal epithelial cell self-renewal following DSS-induced experimental colitis. Sci Rep 2020; 10:10180. [PMID: 32576895 PMCID: PMC7311394 DOI: 10.1038/s41598-020-67152-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 06/04/2020] [Indexed: 02/06/2023] Open
Abstract
CD47 deficient mice are resistant to dextran sulfate sodium (DSS)-induced experimental colitis. The underlying mechanism, however, remains incompletely understood. In this study, we characterized the role of CD47 in modulating homeostasis of gastrointestinal tract. We found that CD47 expression in both human and mouse intestinal epithelium was upregulated in colitic condition compared to that under normal condition. In line with this, CD47 deficiency protected mice from DSS-induced colitis. Analysis based on both intestinal organoid and cultured cell assays showed that CD47 deficiency accelerated intestinal epithelial cell proliferation and migration. Mechanistically, western blot and functional assays indicated that CD47 deficiency promoting mouse intestinal epithelial cell proliferation and migration follow cell injury is likely through upregulating expression of four Yamanaka transcriptional factors Oct4, Sox2, Klf4 and c-Myc (OSKM in abbreviation). Our studies thus reveal CD47 as a negative regulator in intestinal epithelial cell renewal during colitis through downregulating OSKM transcriptional factors.
Collapse
Affiliation(s)
- Yueqin He
- Nanjing University Advanced Institute of Life Sciences, Nanjing, China
| | - Xinlei Sun
- Nanjing University Advanced Institute of Life Sciences, Nanjing, China
| | - Weiwei Rong
- Nanjing University Advanced Institute of Life Sciences, Nanjing, China
| | - Rong Yang
- Nanjing University Advanced Institute of Life Sciences, Nanjing, China
| | - Hongwei Liang
- Nanjing University Advanced Institute of Life Sciences, Nanjing, China
| | - Ying Qi
- Department of Gastroenterology and Hepatology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Limin Li
- Nanjing University Advanced Institute of Life Sciences, Nanjing, China.
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, Nanjing University, Nanjing, Jiangsu, 210093, China.
| | - Ke Zen
- Nanjing University Advanced Institute of Life Sciences, Nanjing, China.
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, Nanjing University, Nanjing, Jiangsu, 210093, China.
| |
Collapse
|
13
|
Cao D, Chen MK, Zhang QF, Zhou YF, Zhang MY, Mai SJ, Zhang YJ, Chen MS, Li XX, Wang HY. Identification of immunological subtypes of hepatocellular carcinoma with expression profiling of immune-modulating genes. Aging (Albany NY) 2020; 12:12187-12205. [PMID: 32544882 PMCID: PMC7343492 DOI: 10.18632/aging.103395] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/20/2020] [Indexed: 12/24/2022]
Abstract
Recent studies demonstrate that immune checkpoint inhibitor (ICI) therapy has achieved success in many types of advanced cancers including advanced hepatocellular carcinoma (HCC). However, ICI therapy is beneficial in only some HCC patients, suggesting that immune-responses are highly variable in HCCs. Therefore, understanding the immune status in HCC microenvironment will facilitate ICI immunotherapy and guide patient selection for the therapy. In this study, we first analyzed the expression profile of immune-modulating genes and their relationship with survival of HCC patients using the data downloaded from The Cancer Genome Atlas - Liver Hepatocellular Carcinoma (TCGA-LIHC) database, and found that the higher expressions of CD276 (B7-H3) and CD47 were significantly associated with poor survival. Then we identified 4 immune subtypes of HCCs with different survivals by using the combination expression of B7-H3 (or CD47) and CD8. Patients with B7-H3low/CD8high or CD47low/CD8high have the best survival while ones with B7-H3high/CD8low or CD47high/CD8low have the worst survival. The 4 immune subtypes were validated in another 72 HCC patients obtained from South China. In conclusion, our findings suggest that HCC patient prognosis is associated with immunophenotypes by T cell infiltration (CD8 expression) and the expression of the adaptive immune resistance gene (B7-H3 or CD47), and this immune classification system will facilitate HCC patient selection for ICI immunotherapy.
Collapse
Affiliation(s)
- Di Cao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Meng-Ke Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Qing-Feng Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yu-Feng Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Mei-Yin Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Shi-Juan Mai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yao-Jun Zhang
- Department of Hepatobiliary Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Min-Shan Chen
- Department of Hepatobiliary Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xiao-Xing Li
- Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hui-Yun Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|
14
|
Chapuy L, Bsat M, Rubio M, Harvey F, Motta V, Schwenter F, Wassef R, Richard C, Deslandres C, Nguyen BN, Soucy G, Hacohen N, Fritz J, Villani AC, Mehta H, Sarfati M. Transcriptomic Analysis and High-dimensional Phenotypic Mapping of Mononuclear Phagocytes in Mesenteric Lymph Nodes Reveal Differences Between Ulcerative Colitis and Crohn's Disease. J Crohns Colitis 2020; 14:393-405. [PMID: 31541232 PMCID: PMC7068244 DOI: 10.1093/ecco-jcc/jjz156] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND AIMS Crohn's disease [CD] and ulcerative colitis [UC] are distinct forms of inflammatory bowel disease. Heterogeneity of HLA-DR+SIRPα + mononuclear phagocytes [MNPs], including macrophages [MΦ], monocyte-derived [Mono] cells, and dendritic cells [DCs], was reported in gut tissue but not yet investigated in mesenteric lymph nodes [MLNs] of IBD patients. We here compared the phenotype, function, and molecular profile of HLA-DR+SIRPα + MNPs in CD and UC MLNs. METHODS Cell distribution, morphology, immune function, and transcriptomic [bulk RNAseq] and high-dimensional protein expression profiles [CyTOF] of HLA-DR+SIRPα + MNPs were examined in MLNs of UC [n = 14], CD [n = 35], and non-IBD [n = 12] patients. RESULTS Elevated frequencies of CD14+CD64+CD163+ [Mono/MΦ-like] MNPs displaying monocyte/MΦ morphology and phagocytic function were a distinct feature of UC MLNs. In CD, the proportion of CD14-CD64-CD163- [DC-like] cells was augmented relative to Mono/MΦ-like cells; DC-like cells drove naïve T cell proliferation, Th1 polarisation, and Th17 TCM plasticity. Gene expression profile corroborated the nature of DC-like cells, best represented by BTLA, SERPINF, IGJ and, of Mono/MΦ-like cells, defined by CD163, MARCO, MAFB, CD300E, S100A9 expression. CyTOF analysis showed that CD123+ plasmacytoid cells predominated over conventional DCs in DC-like cells. Four CD163+ clusters were revealed in Mono/MΦ-like cells, two of which were enriched in MARCO-CD68dimHLA-DRdim monocyte-like cells and MARCOhiCD68hiHLA-DRhi Mɸ, whose proportion increased in UC relative to CD. CONCLUSIONS Defining the landscape of MNPs in MLNs provided evidence for expansion of CD163+ Mono/MΦ-like cells in UC only, highlighting a distinction between UC and CD, and thus the potential contribution of monocyte-like cells in driving colitis.
Collapse
Affiliation(s)
- Laurence Chapuy
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l’Université de Montréal [CRCHUM], Montréal, QC, Canada
| | - Marwa Bsat
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l’Université de Montréal [CRCHUM], Montréal, QC, Canada
| | - Manuel Rubio
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l’Université de Montréal [CRCHUM], Montréal, QC, Canada
| | - François Harvey
- Department of Biomedical Informatics, Centre de Recherche du Centre Hospitalier de l’Université de Montréal [CRCHUM], Montréal, QC, Canada
| | - Vinicius Motta
- McGill Goodman Research Center, McGill University, Montréal, QC, Canada
| | - Frank Schwenter
- Digestive Surgery Department, Centre Hospitalier de l’Université de Montréal [CHUM], Montréal, QC, Canada
| | - Ramses Wassef
- Digestive Surgery Department, Centre Hospitalier de l’Université de Montréal [CHUM], Montréal, QC, Canada
| | - Carole Richard
- Digestive Surgery Department, Centre Hospitalier de l’Université de Montréal [CHUM], Montréal, QC, Canada
| | - Colette Deslandres
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, CHU Sainte-Justine, Université de Montreal, QC, Canada
| | - Bich N Nguyen
- Pathology Department, Centre Hospitalier de l’Université de Montréal [CHUM], Montréal, QC, Canada
| | - Geneviève Soucy
- Pathology Department, Centre Hospitalier de l’Université de Montréal [CHUM], Montréal, QC, Canada
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Jorge Fritz
- Department of Microbiology and Immunology, McGill University, Montréal, Qc, Canada
| | - Alexandra-Chloé Villani
- Broad Institute of MIT and Harvard, Cambridge, MA USA
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Heena Mehta
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l’Université de Montréal [CRCHUM], Montréal, QC, Canada
| | - Marika Sarfati
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l’Université de Montréal [CRCHUM], Montréal, QC, Canada
| |
Collapse
|
15
|
Immune checkpoint molecules. Possible future therapeutic implications in autoimmune diseases. J Autoimmun 2019; 104:102333. [DOI: 10.1016/j.jaut.2019.102333] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023]
|
16
|
Chapuy L, Bsat M, Sarkizova S, Rubio M, Therrien A, Wassef E, Bouin M, Orlicka K, Weber A, Hacohen N, Villani AC, Sarfati M. Two distinct colonic CD14 + subsets characterized by single-cell RNA profiling in Crohn's disease. Mucosal Immunol 2019; 12:703-719. [PMID: 30670762 DOI: 10.1038/s41385-018-0126-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/05/2018] [Accepted: 12/11/2018] [Indexed: 02/04/2023]
Abstract
Inflammatory bowel diseases are associated with dysregulated immune responses in the intestinal tissue. Four molecularly identified macrophage subsets control immune homeostasis in healthy gut. However, the specific roles and transcriptomic profiles of the phenotypically heterogeneous CD14+ macrophage-like population in inflamed gut remain to be investigated in Crohn's disease (CD). Here we identified two phenotypically, morphologically and functionally distinct colonic HLADR+SIRPα+CD14+ subpopulations that were further characterized using single-cell RNA-sequencing (scRNAseq) in CD. Frequencies of CD64hiCD163-/dim cells selectively augmented in inflamed colon and correlated with endoscopic score of disease severity. IL-1β and IL-23-producing CD64hiCD163-/dim cells predominated over TNF-α-producing CD64hiCD163hi cells in lesions. Purified "inflammatory monocyte-like" CD163-, but not "macrophage-like" CD163hi cells, through IL-1β, promoted Th17/Th1 but not Th1 responses in tissue memory CD4+T cells. Unsupervised scRNAseq analysis that captures the entire HLADR+SIRPα+ population revealed six clusters, two of which were enriched in either CD163- or CD163hi cells, and best defined by TREM1/FCAR/FCN1/IL1RN or CD209/MERTK/MRCI/CD163L1 genes, respectively. Selected newly identified discriminating markers were used beyond CD163 to isolate cells that shared pro-Th17/Th1 function with CD163- cells. In conclusion, a molecularly distinct pro-inflammatory CD14+ subpopulation accumulates in inflamed colon, drives intestinal inflammatory T-cell responses, and thus, might contribute to CD disease severity.
Collapse
Affiliation(s)
- Laurence Chapuy
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC, Canada
| | - Marwa Bsat
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC, Canada
| | - Siranush Sarkizova
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Manuel Rubio
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC, Canada
| | - Amélie Therrien
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC, Canada.,Centre Hospitalier de l'Université de Montréal (CHUM), Université de Montréal, Montréal, QC, Canada
| | - Evelyne Wassef
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC, Canada
| | - Mickael Bouin
- Centre Hospitalier de l'Université de Montréal (CHUM), Université de Montréal, Montréal, QC, Canada
| | - Katarzina Orlicka
- Centre Hospitalier de l'Université de Montréal (CHUM), Université de Montréal, Montréal, QC, Canada
| | - Audrey Weber
- Centre Hospitalier de l'Université de Montréal (CHUM), Université de Montréal, Montréal, QC, Canada
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alexandra-Chloé Villani
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Marika Sarfati
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
17
|
Aoki R, Aoki-Yoshida A, Suzuki C, Takayama Y. Indole-3-Pyruvic Acid, an Aryl Hydrocarbon Receptor Activator, Suppresses Experimental Colitis in Mice. THE JOURNAL OF IMMUNOLOGY 2018; 201:3683-3693. [PMID: 30429284 DOI: 10.4049/jimmunol.1701734] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 10/15/2018] [Indexed: 12/18/2022]
Abstract
Aryl hydrocarbon receptor (AHR) agonists are promising immunomodulators that potentially maintain immune tolerance. In this study, we examined the ability of indole-3-pyruvic acid (IPA), a major precursor of microbiota-derived AHR agonists and a proagonist of AHR, to activate AHR. The anti-inflammatory effects of IPA were also evaluated in a mouse model of colitis in comparison with other aromatic pyruvic acids (phenylpyruvic acid and 4-hydroxyphenylpyruvic acid). Among them, IPA showed the strongest ability to activate AHR in vitro and in vivo, and only IPA improved chronic inflammation in an experimental colitis model. IPA attenuated the expression of genes encoding Th1 cytokines and enhanced Il-10 gene expression in the colon. Oral administration of IPA decreased the frequency of IFN-γ+ IL-10- CD4+ T cells and increased that of IFN-γ- IL-10+ CD4+ T cells in the colon lamina propria in a T cell-mediated colitis model. IPA directly promoted the differentiation of type 1 regulatory T cells in vitro. Furthermore, IPA administration attenuated the ability of dendritic cells (DCs) in the mesenteric lymph nodes (MLN) to induce IFN-γ-producing T cells, increased the frequency of CD103+ CD11b- DCs, and decreased the frequency of CD103- CD11b+ DCs in the MLN. Adoptive transfer of MLN CD103+ CD11b- DCs significantly improved the severity of colon inflammation. Treatment with an AHR antagonist inhibited IPA-induced differentiation of type 1 regulatory T cells and the IPA-induced increase in CD103+ CD11b- DCs and attenuated the anti-inflammatory effect of IPA. These findings suggest that IPA potently prevents chronic inflammation in the colon by activating AHR.
Collapse
Affiliation(s)
- Reiji Aoki
- Functional Biomolecules Research Unit, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, Ibaraki 305-0901, Japan; and
| | - Ayako Aoki-Yoshida
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Chise Suzuki
- Functional Biomolecules Research Unit, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, Ibaraki 305-0901, Japan; and
| | - Yoshiharu Takayama
- Functional Biomolecules Research Unit, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, Ibaraki 305-0901, Japan; and
| |
Collapse
|
18
|
Ye X, Wang X, Lu R, Zhang J, Chen X, Zhou G. CD47 as a potential prognostic marker for oral leukoplakia and oral squamous cell carcinoma. Oncol Lett 2018; 15:9075-9080. [PMID: 29805639 PMCID: PMC5958773 DOI: 10.3892/ol.2018.8520] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 03/03/2017] [Indexed: 12/29/2022] Open
Abstract
Cluster of differentiation (CD)47, which acts as a negative indicator for phagocytic cells, is overexpressed on the surface of multiple human solid tumor cell types. Avoiding phagocytosis by CD47 is required for the progression of solid tumors. The present study investigated the expression of CD47 in oral leukoplakia (OLK) and oral squamous cell carcinoma (OSCC), and preliminarily explored the impact of CD47 on the proliferation of OSCC cells. A total of 56 tissue samples, including 36 cases of OLK, 10 cases of OSCC and 10 cases of normal oral mucosa (NOM) were selected to detect the expression of CD47 by immunohistochemistry. For subgroup analysis, OLK samples were divided into OLK with low-risk dysplasia (LR-OLK) and OLK with high-risk dysplasia (HR-OLK). The subcellular localization of CD47 was determined by immunofluorescence in three OSCC cell lines (Tca8113, SCC-9 and Cal-27). The effect of CD47 antibody on the proliferation of the Cal-27 cell line was analyzed using the Cell Counting kit-8 assay. CD47 expression in OLK and OSCC lesions was significantly higher than in NOM (P<0.05). Compared with LR-OLK, the expression of CD47 in HR-OLK and OSCC cells was upregulated (P=0.0327 and P=0.0048, respectively). CD47 was highly expressed in OSCC cell lines (Tca8113, Cal-27 and SCC-9) and weakly expressed in normal oral keratinocytes. The proliferation of Cal-27 cells was inhibited by CD47 antibody in a concentration and time-dependent manner. CD47 may be a reliable biomarker for predicting the progression of oral precancer and cancer, and it may serve as an important molecular target for designing a novel therapy for oral cancer.
Collapse
Affiliation(s)
- Xiaojing Ye
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P.R. China
| | - Xiaojun Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P.R. China
| | - Rui Lu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P.R. China.,Department of Oral Medicine, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P.R. China
| | - Jing Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P.R. China.,Department of Oral Medicine, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P.R. China
| | - Xinming Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P.R. China.,Department of Oral Pathology, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P.R. China
| | - Gang Zhou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P.R. China.,Department of Oral Medicine, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P.R. China
| |
Collapse
|
19
|
Fuke N, Takagi T, Higashimura Y, Tsuji T, Umeda R, Mizushima K, Tanaka M, Suganuma H, Aizawa K, Yajima N, Naito Y. Lactobacillus brevis KB290 With Vitamin A Ameliorates Murine Intestinal Inflammation Associated With the Increase of CD11c+ Macrophage/CD103- Dendritic Cell Ratio. Inflamm Bowel Dis 2018; 24:317-331. [PMID: 29361084 DOI: 10.1093/ibd/izx003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND The ratio of colonic anti-inflammatory CD11c+ macrophages (MPs) to inflammatory CD103- dendritic cells (DCs) plays pivotal roles in intestinal inflammation. Little is known about how the ratio is regulated by lactic acid bacteria (LAB) and bifidobacteria (Bif). We investigated the contribution of LAB/Bif to this ratio. METHODS We established an in vitro experimental system using human myeloblastic KG-1 cells, which differentiate into CD11c+ MP-like (CD11c+ MPL) and CD103- DC-like (CD103- DCL) cells, and explored effective LAB/Bif strains. The selected strain's effect on the colonic CD11c+ MP/CD103- DC ratio and intestinal inflammation was examined in mice, and the strain's underlying mechanisms were investigated in vitro. RESULTS We screened 19 strains of LAB/Bif, and found that Lactobacillus brevis KB290 (KB290) increased the CD11c+ MPL/CD103- DCL cell ratio only in the presence of a vitamin A (VA) metabolite, retinoic acid (RA). Supplementation of KB290 with VA increased the CD11c+ MP/CD103- DC ratio in healthy mouse and prevented the disruption of the ratio during colitis. Supplementation of KB290 with pro-VA (β-carotene) also increased the ratio in healthy mouse and ameliorated the development of colitis. The ratio was increased by reduction of CD103- DCs (or CD103- DCL cells). Our in vitro data suggested that KB290 induced cell death in CD103- DCL cells in the presence of RA signaling. CONCLUSIONS Supplementation of KB290 with VA increases the colonic CD11c+ MP/CD103- DC ratio associated with the amelioration of murine colitis, suggesting a possible way to control intestinal inflammation by LAB.
Collapse
Affiliation(s)
- Nobuo Fuke
- Nature & Wellness Research Department, Innovation Division, Nasushiobara, Tochigi, Japan
| | - Tomohisa Takagi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto, Japan
| | - Yasuki Higashimura
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto, Japan.,Department of Food Science, Ishikawa Prefectural University, Nonoichi, Ishikawa, Japan
| | - Toshifumi Tsuji
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto, Japan
| | - Ryohei Umeda
- Nature & Wellness Research Department, Innovation Division, Nasushiobara, Tochigi, Japan
| | - Katsura Mizushima
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto, Japan
| | - Makoto Tanaka
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto, Japan
| | - Hiroyuki Suganuma
- Nature & Wellness Research Department, Innovation Division, Nasushiobara, Tochigi, Japan
| | - Koichi Aizawa
- Nature & Wellness Research Department, Innovation Division, Nasushiobara, Tochigi, Japan
| | - Nobuhiro Yajima
- Nature & Wellness Research Department, Innovation Division, Nasushiobara, Tochigi, Japan.,Department of Food and Nutritional Science, Tokyo University of Agriculture, Setagaya-ku, Japan
| | - Yuji Naito
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto, Japan
| |
Collapse
|
20
|
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: 409] [Impact Index Per Article: 51.1] [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
|
21
|
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: 95] [Impact Index Per Article: 11.9] [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
|
22
|
Mehta H, Goulet PO, Nguyen V, Pérez G, Koenig M, Senécal JL, Sarfati M. Topoisomerase I peptide-loaded dendritic cells induce autoantibody response as well as skin and lung fibrosis. Autoimmunity 2016; 49:503-513. [DOI: 10.1080/08916934.2016.1230848] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
| | | | | | - Gemma Pérez
- Laboratory for Research in Autoimmunity, Center de Recherche du Center Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Martial Koenig
- Laboratory for Research in Autoimmunity, Center de Recherche du Center Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Jean-Luc Senécal
- Laboratory for Research in Autoimmunity, Center de Recherche du Center Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | | |
Collapse
|
23
|
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: 106] [Impact Index Per Article: 11.8] [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
|
24
|
Gao Q, Zhang Y, Han C, Hu X, Zhang H, Xu X, Tian J, Liu Y, Ding Y, Liu J, Wang C, Guo Z, Yang Y, Cao X. Blockade of CD47 ameliorates autoimmune inflammation in CNS by suppressing IL-1-triggered infiltration of pathogenic Th17 cells. J Autoimmun 2016; 69:74-85. [PMID: 26994903 DOI: 10.1016/j.jaut.2016.03.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/04/2016] [Accepted: 03/06/2016] [Indexed: 12/11/2022]
Abstract
The migration of Th17 cells into central nervous system (CNS) tissue is the key pathogenic step in experimental autoimmune encephalomyelitis (EAE) model. However, the mechanism underlying the pathogenic Th17 cell migration remains elusive. Here we report that blockade of CD47 with CD47-Fc fusion protein is effective in preventing and curing EAE by impairing infiltration of Th17 cells into CNS. However, CD47 deficiency does not directly impair the migration of Th17 cells. Mechanistic studies showed that CD47 deficiency inhibited degradation of inducible nitric oxide synthase (iNOS) in proteasome of macrophages by Src activation and led to the increased nitric oxide (NO) production. Then NO suppressed inflammasome activation-induced IL-1β production. This lower IL-1β reduces the expression of IL-1R1 and migration-related chemokine receptors on CD47(-/-) Th17 cells, inhibiting the ability of Th17 cells to infiltrate into the CNS of CD47(-/-) mice and therefore suppressing EAE development. In vivo administration of exogenous IL-1β indeed promoted the infiltration CD47(-/-) Th17 cells into CNS and antagonized the protective role of CD47 deficiency in EAE pathogenesis. Our results demonstrate a potential preventive and therapeutic application of CD47 blockade in controlling EAE development.
Collapse
Affiliation(s)
- Qiangguo Gao
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China; Department of Cell Biology, Third Military Medical University, Chongqing, 400038, China.
| | - Yi Zhang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Chaofeng Han
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Xiang Hu
- National Key Laboratory of Medical Molecular Biology & Department of Immunology, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Hua Zhang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Xiongfei Xu
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Jun Tian
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yiqi Liu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuanyuan Ding
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Juan Liu
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Chunmei Wang
- National Key Laboratory of Medical Molecular Biology & Department of Immunology, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Zhenhong Guo
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Yongguang Yang
- First Hospital of Jilin University, Changchun, 130012, China
| | - Xuetao Cao
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China; National Key Laboratory of Medical Molecular Biology & Department of Immunology, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China; Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
25
|
van Rees DJ, Szilagyi K, Kuijpers TW, Matlung HL, van den Berg TK. Immunoreceptors on neutrophils. Semin Immunol 2016; 28:94-108. [PMID: 26976825 PMCID: PMC7129252 DOI: 10.1016/j.smim.2016.02.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 02/24/2016] [Accepted: 02/26/2016] [Indexed: 12/12/2022]
Abstract
Neutrophil activities must be tightly controlled to maintain immune homeostasis. Activating and inhibitory receptors balance the outcome of immune cell activation. Immunoreceptors contain Ig-like extracellular domains and signal via ITAMs or ITIMs. Syk or SHP/SHIP mediate downstream signaling after immunoreceptor activation. Targeting immunoreceptors provides opportunities for therapeutic interventions.
Neutrophils play a critical role in the host defense against infection, and they are able to perform a variety of effector mechanisms for this purpose. However, there are also a number of pathological conditions, including autoimmunity and cancer, in which the activities of neutrophils can be harmful to the host. Thus the activities of neutrophils need to be tightly controlled. As in the case of other immune cells, many of the neutrophil effector functions are regulated by a series of immunoreceptors on the plasma membrane. Here, we review what is currently known about the functions of the various individual immunoreceptors and their signaling in neutrophils. While these immunoreceptors allow for the recognition of a diverse range of extracellular ligands, such as cell surface structures (like proteins, glycans and lipids) and extracellular matrix components, they commonly signal via conserved ITAM or ITIM motifs and their associated downstream pathways that depend on the phosphorylation of tyrosine residues in proteins and/or inositol lipids. This allows for a balanced homeostatic regulation of neutrophil effector functions. Given the number of available immunoreceptors and their fundamental importance for neutrophil behavior, it is perhaps not surprising that pathogens have evolved means to evade immune responses through some of these pathways. Inversely, some of these receptors evolved to specifically recognize these pathogens. Finally, some interactions mediated by immunoreceptors in neutrophils have been identified as promising targets for therapeutic intervention.
Collapse
Affiliation(s)
- Dieke J van Rees
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Katka Szilagyi
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Taco W Kuijpers
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Hanke L Matlung
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Timo K van den Berg
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
26
|
Morishita K, Coimbra R, Langness S, Eliceiri BP, Costantini TW. Neuroenteric axis modulates the balance of regulatory T cells and T-helper 17 cells in the mesenteric lymph node following trauma/hemorrhagic shock. Am J Physiol Gastrointest Liver Physiol 2015; 309:G202-8. [PMID: 26045612 DOI: 10.1152/ajpgi.00097.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/27/2015] [Indexed: 01/31/2023]
Abstract
CD103(+) dendritic cells (DCs) continuously migrate from the intestine to the mesenteric lymph nodes (MLNs) and maintain tolerance by driving the development of regulatory T cells (Treg) in the gut. The relative expression of Treg and T-helper 17 (Th17) cells determines the balance between tolerance and immunity in the gut. We hypothesized that trauma/hemorrhagic shock (T/HS) would decrease the CD103(+) DC population in the mesenteric lymph and alter the Treg-to-Th17 ratio in the MLN. We further hypothesized that vagus nerve stimulation (VNS) would promote tolerance to inflammation by increasing the Treg-to-Th17 ratio in the MLN after injury. Male rats were assigned to sham shock (SS), trauma/sham shock (T/SS), or T/HS. T/HS was induced by laparotomy and 60 min of HS (blood pressure 35 mmHg) followed by fluid resuscitation. A separate cohort of animals underwent cervical VNS after the HS phase. MLN samples were collected 24 h after resuscitation. The CD103(+) DC population and Treg-to-Th17 cell ratio in the MLN were decreased after T/HS compared with SS and T/SS, suggesting a shift to an inflammatory response. VNS prevented the T/HS-induced decrease in the CD103(+) DC population and increased the Treg-to-Th17 ratio compared with T/HS alone. VNS alters the gut inflammatory response to injury by modulating the Treg-Th17 cell balance in the MLN. VNS promotes tolerance to inflammation in the gut, further supporting its ability to modulate the inflammatory set point and alter the response to injury.
Collapse
Affiliation(s)
- Koji Morishita
- Division of Trauma, Surgical Critical Care, Burns, and Acute Care Surgery, Department of Surgery, University of California San Diego Health Sciences, San Diego, California
| | - Raul Coimbra
- Division of Trauma, Surgical Critical Care, Burns, and Acute Care Surgery, Department of Surgery, University of California San Diego Health Sciences, San Diego, California
| | - Simone Langness
- Division of Trauma, Surgical Critical Care, Burns, and Acute Care Surgery, Department of Surgery, University of California San Diego Health Sciences, San Diego, California
| | - Brian P Eliceiri
- Division of Trauma, Surgical Critical Care, Burns, and Acute Care Surgery, Department of Surgery, University of California San Diego Health Sciences, San Diego, California
| | - Todd W Costantini
- Division of Trauma, Surgical Critical Care, Burns, and Acute Care Surgery, Department of Surgery, University of California San Diego Health Sciences, San Diego, California
| |
Collapse
|
27
|
Shi L, Bian Z, Chen CXJ, Guo YN, Lv Z, Zeng C, Liu Z, Zen K, Liu Y. CD47 deficiency ameliorates autoimmune nephritis in Fas(lpr) mice by suppressing IgG autoantibody production. J Pathol 2015; 237:285-95. [PMID: 26095930 DOI: 10.1002/path.4574] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 05/11/2015] [Accepted: 06/08/2015] [Indexed: 11/07/2022]
Abstract
CD47, a self-recognition marker, plays an important role in both innate and adaptive immune responses. To explore the potential role of CD47 in activation of autoreactive T and B cells and the production of autoantibodies in autoimmune disease, especially systemic lupus erythematosus (SLE), we have generated CD47 knockout Fas(lpr) (CD47(-/-) -Fas(lpr) ) mice and examined histopathological changes in the kidneys, cumulative survival rates, proteinuria, extent of splenomegaly and autoantibodies, serum chemistry and immunological parameters. In comparison with Fas(lpr) mice, CD47(-/-) -Fas(lpr) mice exhibit a prolonged lifespan and delayed autoimmune nephritis, including glomerular cell proliferation, basement membrane thickening, acute tubular atrophy and vacuolization. CD47(-/-) -Fas(lpr) mice have lower levels of proteinuria, associated with reduced deposition of complement C3 and C1q, and IgG but not IgM in the glomeruli, compared to age-matched Fas(lpr) mice. Serum levels of antinuclear antibodies and anti-double-stranded DNA antibodies are significantly lower in CD47(-/-) -Fas(lpr) than in Fas(lpr) mice. CD47(-/-) -Fas(lpr) mice also display less pronounced splenomegaly than Fas(lpr) mice. The mechanistic studies further suggest that CD47 deficiency impairs the antigenic challenge-induced production of IgG but not IgM, and that this effect is associated with reduction of T follicular cells and impairment of germinal centre development in lymphoid tissues. In conclusion, our results demonstrate that CD47 deficiency ameliorates lupus nephritis in Fas(lpr) mice via suppression of IgG autoantibody production.
Collapse
Affiliation(s)
- Lei Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Centre for MicroRNA Biology and Biotechnology, Nanjing University School of Life Sciences, Peoples Republic of China
- Center for Diagnostics and Therapeutics, Center for Inflammation, Immunity and Infection, Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Zhen Bian
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Centre for MicroRNA Biology and Biotechnology, Nanjing University School of Life Sciences, Peoples Republic of China
- Center for Diagnostics and Therapeutics, Center for Inflammation, Immunity and Infection, Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Celia X J Chen
- Center for Diagnostics and Therapeutics, Center for Inflammation, Immunity and Infection, Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Ya-Nan Guo
- Center for Diagnostics and Therapeutics, Center for Inflammation, Immunity and Infection, Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Zhiyuan Lv
- Center for Diagnostics and Therapeutics, Center for Inflammation, Immunity and Infection, Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Caihong Zeng
- National Clinical Research Centre for Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, People's Republic of China
| | - Zhihong Liu
- National Clinical Research Centre for Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, People's Republic of China
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Centre for MicroRNA Biology and Biotechnology, Nanjing University School of Life Sciences, Peoples Republic of China
- Center for Diagnostics and Therapeutics, Center for Inflammation, Immunity and Infection, Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Yuan Liu
- Center for Diagnostics and Therapeutics, Center for Inflammation, Immunity and Infection, Department of Biology, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
28
|
Navarathna DHMLP, Stein EV, Lessey-Morillon EC, Nayak D, Martin-Manso G, Roberts DD. CD47 Promotes Protective Innate and Adaptive Immunity in a Mouse Model of Disseminated Candidiasis. PLoS One 2015; 10:e0128220. [PMID: 26010544 PMCID: PMC4444371 DOI: 10.1371/journal.pone.0128220] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 04/24/2015] [Indexed: 11/19/2022] Open
Abstract
CD47 is a widely expressed receptor that regulates immunity by engaging its counter-receptor SIRPα on phagocytes and its secreted ligand thrombospondin-1. Mice lacking CD47 can exhibit enhanced or impaired host responses to bacterial pathogens, but its role in fungal immunity has not been examined. cd47-/- mice on a C57BL/6 background showed significantly increased morbidity and mortality following Candida albicans infection when compared with wild-type mice. Despite normal fungal colonization at earlier times, cd47-/- mice at four days post-infection had increased colonization of brain and kidneys accompanied by stronger inflammatory reactions. Neutrophil and macrophage numbers were significantly elevated in kidneys and neutrophils in the brains of infected cd47-/- mice. However, no defect in phagocytic activity towards C. albicans was observed in cd47-/- bone-marrow-derived macrophages, and neutrophil and macrophage killing of C. albicans was not impaired. CD47-deficiency did not alter the early humoral immune response to C. albicans. Th1, Th2, and Th17 population of CD4+ T cells were expanded in the spleen, and gene expression profiles of spleen and kidney showed stronger pro-inflammatory signaling in infected cd47-/- mice. The chemoattractant chemokines MIP-2α and MIP-2β were highly expressed in infected spleens of cd47-/- mice. G-CSF, GM-CSF, and the inflammasome component NLRP3 were more highly expressed in infected cd47-/- kidneys than in infected wild-type controls. Circulating pro- (TNF-α, IL-6) and anti-inflammatory cytokines (IL-10) were significantly elevated, but IL-17 was decreased. These data indicate that CD47 plays protective roles against disseminated candidiasis and alters pro-inflammatory and immunosuppressive pathways known to regulate innate and T cell immunity.
Collapse
Affiliation(s)
- Dhammika H. M. L. P. Navarathna
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, United States of America
| | - Erica V. Stein
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, United States of America
- Microbiology and Immunology Program of the Institute for Biomedical Sciences, Departments of Microbiology, Immunology and Tropical Medicine, George Washington University, Washington, D.C. 20037, United States of America
| | - Elizabeth C. Lessey-Morillon
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, United States of America
| | - Debasis Nayak
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Gema Martin-Manso
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, United States of America
| | - David D. Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, United States of America
- * E-mail:
| |
Collapse
|
29
|
|
30
|
Scott CL, Tfp ZM, Beckham KSH, Douce G, Mowat AM. Signal regulatory protein alpha (SIRPα) regulates the homeostasis of CD103(+) CD11b(+) DCs in the intestinal lamina propria. Eur J Immunol 2014; 44:3658-68. [PMID: 25236797 PMCID: PMC4284040 DOI: 10.1002/eji.201444859] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 08/14/2014] [Accepted: 09/16/2014] [Indexed: 12/23/2022]
Abstract
Signal regulatory protein alpha (SIRPα/CD172a) is a conserved transmembrane protein thought to play an inhibitory role in immune function by binding the ubiquitous ligand CD47. SIRPα expression has been used to identify dendritic cell subsets across species and here we examined its expression and function on intestinal DCs in mice. Normal mucosa contains four subsets of DCs based on their expression of CD103 and CD11b and three of these express SIRPα. However, loss of SIRPα signaling in mice leads to a selective reduction in the CD103+CD11b+ subset of DCs in the small intestine, colon, and among migratory DCs in the mesenteric lymph node. In parallel, these mice have reduced numbers of TH17 cells in steady-state intestinal mucosa, and a defective TH17 response to Citrobacter infection. Identical results were obtained in CD47KO mice. DC precursors from SIRPα mutant mice had an enhanced ability to generate CD103+CD11b+ DCs in vivo, but CD103+CD11b+ DCs from mutant mice were more prone to die by apoptosis. These data show a previously unappreciated and crucial role for SIRPα in the homeostasis of CD103+CD11b+ DCs in the intestine, as well as providing further evidence that this subset of DCs is critical for the development of mucosal TH17 responses.
Collapse
Affiliation(s)
- Charlotte L Scott
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Veterinary, Medical and Life Sciences, University of Glasgow, Scotland, UK; VIB Ghent University, Inflammation Research Centre (IRC), Laboratory of Immunoregulation, Ghent (Zwijnaarde), Belgium; Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | | | | | | | | |
Collapse
|
31
|
Wong ASL, Mortin-Toth S, Sung M, Canty AJ, Gulban O, Greaves DR, Danska JS. Polymorphism in the innate immune receptor SIRPα controls CD47 binding and autoimmunity in the nonobese diabetic mouse. THE JOURNAL OF IMMUNOLOGY 2014; 193:4833-44. [PMID: 25305319 DOI: 10.4049/jimmunol.1401984] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The signal regulatory protein (SIRP) locus encodes a family of paired receptors that mediate both activating and inhibitory signals and is associated with type 1 diabetes (T1D) risk. The NOD mouse model recapitulates multiple features of human T1D and enables mechanistic analysis of the impact of genetic variations on disease. In this study, we identify Sirpa encoding an inhibitory receptor on myeloid cells as a gene in the insulin-dependent diabetes locus 13.2 (Idd13.2) that drives islet inflammation and T1D. Compared to T1D-resistant strains, the NOD variant of SIRPα displayed greater binding to its ligand CD47, as well as enhanced T cell proliferation and diabetogenic potency. Myeloid cell-restricted expression of a Sirpa transgene accelerated disease in a dose-dependent manner and displayed genetic and functional interaction with the Idd5 locus to potentiate insulitis progression. Our study demonstrates that variations in both SIRPα sequence and expression level modulate T1D immunopathogenesis. Thus, we identify Sirpa as a T1D risk gene and provide insight into the complex mechanisms by which disease-associated variants act in concert to drive defined stages in disease progression.
Collapse
Affiliation(s)
- Andrea Sut Ling Wong
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S1A8, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario M5G1X8, Canada
| | - Steven Mortin-Toth
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario M5G1X8, Canada
| | - Michael Sung
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario M5G1X8, Canada
| | - Angelo J Canty
- Department of Mathematics and Statistics, McMaster University, Hamilton, Ontario L8S4L8, Canada
| | - Omid Gulban
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario M5G1X8, Canada
| | - David R Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX13RE, United Kingdom; and
| | - Jayne S Danska
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S1A8, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario M5G1X8, Canada; Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S1A8, Canada
| |
Collapse
|
32
|
Soto-Pantoja DR, Terabe M, Ghosh A, Ridnour LA, DeGraff WG, Wink DA, Berzofsky JA, Roberts DD. CD47 in the tumor microenvironment limits cooperation between antitumor T-cell immunity and radiotherapy. Cancer Res 2014; 74:6771-83. [PMID: 25297630 DOI: 10.1158/0008-5472.can-14-0037-t] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although significant advances in radiotherapy have increased its effectiveness in many cancer settings, general strategies to widen the therapeutic window between normal tissue toxicity and malignant tumor destruction would still offer great value. CD47 blockade has been found to confer radioprotection to normal tissues while enhancing tumor radiosensitivity. Here, we report that CD47 blockade directly enhances tumor immunosurveillance by CD8(+) T cells. Combining CD47 blockade with irradiation did not affect fibrosarcoma growth in T cell-deficient mice, whereas adoptive transfer of tumor-specific CD8(+) T cells restored combinatorial efficacy. Furthermore, ablation of CD8(+) T cells abolished radiotherapeutic response in immunocompetent syngeneic hosts. CD47 blockade in either target cells or effector cells was sufficient to enhance antigen-dependent CD8(+) CTL-mediated tumor cell killing in vitro. In CD47-deficient syngeneic hosts, engrafted B16 melanomas were 50% more sensitive to irradiation, establishing that CD47 expression in the microenvironment was sufficient to limit tumor radiosensitivity. Mechanistic investigations revealed increased tumor infiltration by cytotoxic CD8(+) T cells in a CD47-deficient microenvironment, with an associated increase in T cell-dependent intratumoral expression of granzyme B. Correspondingly, an inverse correlation between CD8(+) T-cell infiltration and CD47 expression was observed in human melanomas. Our findings establish that blocking CD47 in the context of radiotherapy enhances antitumor immunity by directly stimulating CD8(+) cytotoxic T cells, with the potential to increase curative responses.
Collapse
Affiliation(s)
- David R Soto-Pantoja
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Masaki Terabe
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Arunima Ghosh
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Lisa A Ridnour
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - William G DeGraff
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - David A Wink
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
33
|
Terai T, Osawa S, Tani S, Oishi S, Arai Y, Yamada T, Sugimoto M, Furuta T, Kanaoka S, Miyajima H, Sugimoto K. Induction of murine TNBS colitis is strictly controlled by a modified method using continuous inhalation anesthesia with sevoflurane. Dig Dis Sci 2014; 59:1415-1427. [PMID: 24452840 DOI: 10.1007/s10620-013-3023-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 12/30/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND Trinitrobenzenesulfonic acid (TNBS)-induced colitis is one of the most widely used experimental colitis models. However, there is no standard procedure for inducing colitis by TNBS because it is difficult to achieve a uniform distribution of colitis. We have developed a modified method of murine TNBS-induced colitis that involves inhalation anesthesia with sevoflurane combined with both single and repeated TNBS administrations. AIMS To compare the usefulness of our newly developed method for inducing murine TNBS-induced colitis with that of conventional intraperitoneal anesthesia. METHODS TNBS in ethanol was administered to C57BL/6J mice held in an inverted vertical position either under continuous inhalation anesthesia with sevoflurane, in accordance with our newly developed method, or by intraperitoneal injection with 2.5 % avertin, in accordance with the conventional procedure. Body weight change, cytokine profile, and histological findings were examined during the course of colitis. RESULTS The dispersion of anesthesia time, TNBS retention time, and nadir weight during the course of colitis was decreased using the newly developed method compared with the conventional procedure. Optimization of the modified TNBS-induced colitis, as evidenced by the predominant expression of Th1 and Th17 cytokines on day 7, was attained by the injection of 2.25 mg TNBS in 55 % ethanol. Regulation of the TNBS retention time using inhalation anesthesia with sevoflurane allowed strict control of the disease severity of TNBS-induced colitis. Using the modified method we were also able to develop a chronic TNBS-induced colitis model by repeated TNBS administration without excessive mortality of the mice. CONCLUSIONS Our modified method for murine TNBS-induced colitis using continuous inhalation anesthesia with sevoflurane provides a better experimental colitis model following both single and repeated TNBS administrations.
Collapse
Affiliation(s)
- Tomohiro Terai
- First Department of Medicine, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Wenzel UA, Magnusson MK, Rydström A, Jonstrand C, Hengst J, Johansson MEV, Velcich A, Öhman L, Strid H, Sjövall H, Hansson GC, Wick MJ. Spontaneous colitis in Muc2-deficient mice reflects clinical and cellular features of active ulcerative colitis. PLoS One 2014; 9:e100217. [PMID: 24945909 PMCID: PMC4063762 DOI: 10.1371/journal.pone.0100217] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 05/23/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The colonic mucus layer plays a critical role in intestinal homeostasis by limiting contact between luminal bacteria and the mucosal immune system. A defective mucus barrier in animal models allows bacterial contact with the intestinal epithelium and results in spontaneous colitis. A defective mucus barrier is also a key feature of active ulcerative colitis (UC). Alterations in the immune compartment due to intestinal bacterial breach in mice lacking the colon mucus barrier have not been characterized and correlated to active UC. AIMS To characterize alterations in the immune compartment due to intestinal bacterial breach in Muc2-/- mice, which lack the colon mucus barrier, and correlate the findings to active UC. METHODS Bacterial contact with colon epithelium and penetration into colon tissue was examined in Muc2-/- mice and colon biopsies from patients with active UC using fluorescence microscopy and qPCR. Neutrophils, lymphocytes, CD103+ dendritic cell subsets and macrophages in colon from Muc2-/- mice and biopsies from UC patients were quantitated by flow cytometry. RESULTS Inflamed UC patients and Muc2-/- mice had bacteria in contact with the colon epithelium. Bacterial rRNA was present in colonic mucosa in humans and Muc2-/- mice and in the draining lymph nodes of mice. Inflamed Muc2-/- mice and UC patients had elevated colon neutrophils, T cells and macrophages while a reduced frequency of CD103+ DCs was present in the inflamed colon of both mice and humans. CONCLUSIONS The parallel features of the colon immune cell compartment in Muc2-/- mice and UC patients supports the usefulness of this model to understand the early phase of spontaneous colitis and will provide insight into novel strategies to treat UC.
Collapse
Affiliation(s)
- Ulf A. Wenzel
- Department of Microbiology and Immunology and the Mucosal Immunobiology and Vaccine Center (MIVAC), Institute of Biomedicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maria K. Magnusson
- Department of Microbiology and Immunology and the Mucosal Immunobiology and Vaccine Center (MIVAC), Institute of Biomedicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; and MIVAC, University of Gothenburg, Gothenburg, Sweden
| | - Anna Rydström
- Department of Microbiology and Immunology and the Mucosal Immunobiology and Vaccine Center (MIVAC), Institute of Biomedicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Caroline Jonstrand
- Department of Microbiology and Immunology and the Mucosal Immunobiology and Vaccine Center (MIVAC), Institute of Biomedicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Julia Hengst
- Department of Microbiology and Immunology and the Mucosal Immunobiology and Vaccine Center (MIVAC), Institute of Biomedicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Malin EV. Johansson
- Department of Medical Biochemistry, Institute of Biomedicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; and MIVAC, University of Gothenburg, Gothenburg, Sweden
| | - Anna Velcich
- Department of Oncology, Albert Einstein Cancer Center/Montefiore Medical Center, New York City, New York, United States of America
| | - Lena Öhman
- Department of Microbiology and Immunology and the Mucosal Immunobiology and Vaccine Center (MIVAC), Institute of Biomedicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; and MIVAC, University of Gothenburg, Gothenburg, Sweden
| | - Hans Strid
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; and MIVAC, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Sjövall
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; and MIVAC, University of Gothenburg, Gothenburg, Sweden
| | - Gunnar C. Hansson
- Department of Medical Biochemistry, Institute of Biomedicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; and MIVAC, University of Gothenburg, Gothenburg, Sweden
| | - Mary Jo Wick
- Department of Microbiology and Immunology and the Mucosal Immunobiology and Vaccine Center (MIVAC), Institute of Biomedicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- * E-mail:
| |
Collapse
|
35
|
Haque A, Best SE, Montes de Oca M, James KR, Ammerdorffer A, Edwards CL, de Labastida Rivera F, Amante FH, Bunn PT, Sheel M, Sebina I, Koyama M, Varelias A, Hertzog PJ, Kalinke U, Gun SY, Rénia L, Ruedl C, MacDonald KPA, Hill GR, Engwerda CR. Type I IFN signaling in CD8- DCs impairs Th1-dependent malaria immunity. J Clin Invest 2014; 124:2483-96. [PMID: 24789914 DOI: 10.1172/jci70698] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Many pathogens, including viruses, bacteria, and protozoan parasites, suppress cellular immune responses through activation of type I IFN signaling. Recent evidence suggests that immune suppression and susceptibility to the malaria parasite, Plasmodium, is mediated by type I IFN; however, it is unclear how type I IFN suppresses immunity to blood-stage Plasmodium parasites. During experimental severe malaria, CD4+ Th cell responses are suppressed, and conventional DC (cDC) function is curtailed through unknown mechanisms. Here, we tested the hypothesis that type I IFN signaling directly impairs cDC function during Plasmodium infection in mice. Using cDC-specific IFNAR1-deficient mice, and mixed BM chimeras, we found that type I IFN signaling directly affects cDC function, limiting the ability of cDCs to prime IFN-γ-producing Th1 cells. Although type I IFN signaling modulated all subsets of splenic cDCs, CD8- cDCs were especially susceptible, exhibiting reduced phagocytic and Th1-promoting properties in response to type I IFNs. Additionally, rapid and systemic IFN-α production in response to Plasmodium infection required type I IFN signaling in cDCs themselves, revealing their contribution to a feed-forward cytokine-signaling loop. Together, these data suggest abrogation of type I IFN signaling in CD8- splenic cDCs as an approach for enhancing Th1 responses against Plasmodium and other type I IFN-inducing pathogens.
Collapse
|
36
|
Barclay AN, van den Berg TK. The Interaction Between Signal Regulatory Protein Alpha (SIRPα) and CD47: Structure, Function, and Therapeutic Target. Annu Rev Immunol 2014; 32:25-50. [DOI: 10.1146/annurev-immunol-032713-120142] [Citation(s) in RCA: 448] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- A. Neil Barclay
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK;
| | - Timo K. van den Berg
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, 1066 CX Amsterdam, The Netherlands;
| |
Collapse
|
37
|
Murata Y, Kotani T, Ohnishi H, Matozaki T. The CD47-SIRPα signalling system: its physiological roles and therapeutic application. J Biochem 2014; 155:335-44. [PMID: 24627525 DOI: 10.1093/jb/mvu017] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Signal regulatory protein α (SIRPα), also known as SHPS-1/BIT/ CD172a, is an immunoglobulin superfamily protein that binds to the protein tyrosine phosphatases SHP-1 and SHP-2 through its cytoplasmic region. CD47, another immunoglobulin superfamily protein, is a ligand for SIRPα, with the two proteins constituting a cell-cell communication system (the CD47-SIRPα signalling system). SIRPα is particularly abundant in the myeloid-lineage hematopoietic cells such as macrophages or dendritic cells (DCs), whereas CD47 is expressed ubiquitously. Interaction of CD47 (on red blood cells) with SIRPα (on macrophages) is thought to prevent the phagocytosis by the latter cells of the former cells, determining the lifespan of red blood cells. Recent studies further indicate that this signalling system plays important roles in engraftment of hematopoietic stem cells as well as in tumour immune surveillance through regulation of the phagocytic activity of macrophages. In the immune system, the CD47-SIRPα interaction is also important for the development of a subset of CD11c(+)DCs as well as organization of secondary lymphoid organs. Finally, the CD47-SIRPα signalling system likely regulates bone homeostasis by osteoclast development. Newly emerged functions of the CD47-SIRPα signalling system thus provide multiple therapeutic strategies for cancer, autoimmune diseases and bone disorders.
Collapse
Affiliation(s)
- Yoji Murata
- Department of Biochemistry and Molecular Biology, Division of Molecular and Cellular Signaling, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan; and Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-Machi, Maebashi, Gunma 371-8514, Japan
| | - Takenori Kotani
- Department of Biochemistry and Molecular Biology, Division of Molecular and Cellular Signaling, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan; and Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-Machi, Maebashi, Gunma 371-8514, Japan
| | - Hiroshi Ohnishi
- Department of Biochemistry and Molecular Biology, Division of Molecular and Cellular Signaling, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan; and Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-Machi, Maebashi, Gunma 371-8514, Japan
| | - Takashi Matozaki
- Department of Biochemistry and Molecular Biology, Division of Molecular and Cellular Signaling, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan; and Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-Machi, Maebashi, Gunma 371-8514, Japan
| |
Collapse
|
38
|
Osteopontin expression by CD103- dendritic cells drives intestinal inflammation. Proc Natl Acad Sci U S A 2014; 111:E856-65. [PMID: 24550510 DOI: 10.1073/pnas.1316447111] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Intestinal CD103(-) dendritic cells (DCs) are pathogenic for colitis. Unveiling molecular mechanisms that render these cells proinflammatory is important for the design of specific immunotherapies. In this report, we demonstrated that mesenteric lymph node CD103(-) DCs express, among other proinflammatory cytokines, high levels of osteopontin (Opn) during experimental colitis. Opn expression by CD103(-) DCs was crucial for their immune profile and pathogenicity, including induction of T helper (Th) 1 and Th17 cell responses. Adoptive transfer of Opn-deficient CD103(-) DCs resulted in attenuated colitis in comparison to transfer of WT CD103(-) DCs, whereas transgenic CD103(-) DCs that overexpress Opn were highly pathogenic in vivo. Neutralization of secreted Opn expressed exclusively by CD103(-) DCs restrained disease severity. Also, Opn deficiency resulted in milder disease, whereas systemic neutralization of secreted Opn was therapeutic. We determined a specific domain of the Opn protein responsible for its CD103(-) DC-mediated proinflammatory effect. We demonstrated that disrupting the interaction of this Opn domain with integrin α9, overexpressed on colitic CD103(-) DCs, suppressed the inflammatory potential of these cells in vitro and in vivo. These results add unique insight into the biology of CD103(-) DCs and their function during inflammatory bowel disease.
Collapse
|
39
|
Abstract
Intestinal epithelial cells were once thought to be inert, non-responsive cells that simply acted as a physical barrier that prevents the contents of the intestinal lumen from accessing the underlying tissue. However, it is now clear that these cells express a full repertoire of Toll- and Nod-like receptors, and that their activation by components of the microbiota is vital for the development of a functional epithelium, maintenance of barrier integrity, and defense against pathogenic organisms. Additionally, mounting evidence suggests that epithelial sensing of bacteria plays a significant role in the management of the numbers and types of microbes present in the gut microbiota via the production of antimicrobial peptides and other microbe-modulatory products. This is a critical process, as it is now becoming apparent that alterations in the composition of the microbiota can predispose an individual to a wide variety of chronic diseases. In this review, we will discuss the bacterial pattern recognition receptors that are known to be expressed by the intestinal epithelium, and how each of them individually contributes to these vital protective functions. Moreover, we will review what is known about the communication between epithelial cells and various classes of underlying leukocytes, and discuss how they interact with the microbiota to form a three-part relationship that maintains homeostasis in the gut.
Collapse
|
40
|
Azcutia V, Routledge M, Williams MR, Newton G, Frazier WA, Manica A, Croce KJ, Parkos CA, Schmider AB, Turman MV, Soberman RJ, Luscinskas FW. CD47 plays a critical role in T-cell recruitment by regulation of LFA-1 and VLA-4 integrin adhesive functions. Mol Biol Cell 2013; 24:3358-68. [PMID: 24006483 PMCID: PMC3814154 DOI: 10.1091/mbc.e13-01-0063] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
CD47 plays an important but incompletely understood role in the innate and adaptive immune responses. CD47, also called integrin-associated protein, has been demonstrated to associate in cis with β1 and β3 integrins. Here we test the hypothesis that CD47 regulates adhesive functions of T-cell α4β1 (VLA-4) and αLβ2 (LFA-1) in in vivo and in vitro models of inflammation. Intravital microscopy studies reveal that CD47(-/-) Th1 cells exhibit reduced interactions with wild-type (WT) inflamed cremaster muscle microvessels. Similarly, murine CD47(-/-) Th1 cells, as compared with WT, showed defects in adhesion and transmigration across tumor necrosis factor-α (TNF-α)-activated murine endothelium and in adhesion to immobilized intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion protein 1 (VCAM-1) under flow conditions. Human Jurkat T-cells lacking CD47 also showed reduced adhesion to TNF-α-activated endothelium and ICAM-1 and VCAM-1. In cis interactions between Jurkat T-cell β2 integrins and CD47 were detected by fluorescence lifetime imaging microscopy. Unexpectedly, Jurkat CD47 null cells exhibited a striking defect in β1 and β2 integrin activation in response to Mn(2+) or Mg(2+)/ethylene glycol tetraacetic acid treatment. Our results demonstrate that CD47 associates with β2 integrins and is necessary to induce high-affinity conformations of LFA-1 and VLA-4 that recognize their endothelial cell ligands and support leukocyte adhesion and transendothelial migration.
Collapse
Affiliation(s)
- Veronica Azcutia
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115 Harvard Medical School, Boston, MA 02115 Department of Biochemistry and Molecular Biophysics, Washington University, St. Louis, MO 63130 Instituto de Cardiologia do Rio Grande do Sul, Fundação Universitária de Cardiologia, Porto Alegre 90010-395, Brazil Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115 Division of Gastrointestinal Pathology, Emory University School of Medicine, Atlanta, GA 30322 Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Martinelli R, Newton G, Carman CV, Greenwood J, Luscinskas FW. Novel role of CD47 in rat microvascular endothelium: signaling and regulation of T-cell transendothelial migration. Arterioscler Thromb Vasc Biol 2013; 33:2566-76. [PMID: 23990210 DOI: 10.1161/atvbaha.113.301903] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Although endothelial CD47, a member of the immunoglobulin superfamily, has been implicated in leukocyte diapedesis, its capacity for intracellular signaling and physical localization during this process has not been addressed in detail. This study examined endothelial CD47 spatiotemporal behavior and signaling pathways involved in regulating T-cell transendothelial migration. APPROACH AND RESULTS By biochemical methods, transmigration assays, and live-cell microscopy techniques, we show that endothelial CD47 engagement results in intracellular calcium mobilization, increased permeability, and activation of Src and AKT1/phosphoinositide 3-kinase in brain microvascular endothelial cells. These signaling pathways converge to induce cytoskeleton remodeling and vascular endothelial cadherin phosphorylation, which are necessary steps during T-cell transendothelial migration. In addition, during T-cell migration, transmigratory cups and podo-prints enriched in CD47 appear on the surface of the endothelium, indicating that the spatial distribution of CD47 changes after its engagement. Consistent with previous findings of intercellular adhesion molecule 1, blockade of CD47 results in decreased T-cell transmigration across microvascular endothelium. The overlapping effect of intercellular adhesion molecule 1 and CD47 suggests their involvement in different steps of the diapedesis process. CONCLUSIONS These data reveal a novel role for CD47-mediated signaling in the control of the molecular network governing endothelial-dependent T-cell diapedesis.
Collapse
Affiliation(s)
- Roberta Martinelli
- From the Department of Pathology, Center for Excellence in Vascular Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (R.M., G.N., F.W.L.); Department of Medicine, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (R.M., C.V.C.); and Department of Cell Biology, Institute of Ophthalmology, UCL, London, United Kingdom (R.M., J.G.)
| | | | | | | | | |
Collapse
|
42
|
Miller TW, Kaur S, Ivins-O’Keefe K, Roberts DD. Thrombospondin-1 is a CD47-dependent endogenous inhibitor of hydrogen sulfide signaling in T cell activation. Matrix Biol 2013; 32:316-24. [PMID: 23499828 PMCID: PMC3706541 DOI: 10.1016/j.matbio.2013.02.009] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 02/15/2013] [Accepted: 02/28/2013] [Indexed: 01/17/2023]
Abstract
Thrombospondin-1 is a potent suppressor of T cell activation via its receptor CD47. However, the precise mechanism for this inhibition remains unclear. Because H2S is an endogenous potentiator of T cell activation and is necessary for full T cell activation, we hypothesized that thrombospondin-1 signaling through CD47 inhibits T cell activation by antagonizing H2S signaling. Primary T cells from thrombospondin-1 null mice were more sensitive to H2S-dependent activation assessed by proliferation and induction of interleukin-2 and CD69 mRNAs. Exogenous thrombospondin-1 inhibited H2S responses in wild type and thrombospondin-1 null T cells but enhanced the same responses in CD47 null T cells. Fibronectin, which shares integrin and glycosaminoglycan binding properties with thrombospondin-1 but not CD47 binding, did not inhibit H2S signaling. A CD47-binding peptide derived from thrombospondin-1 inhibited H2S-induced activation, whereas two other functional sequences from thrombospondin-1 enhanced H2S signaling. Therefore, engaging CD47 is necessary and sufficient for thrombospondin-1 to inhibit H2S-dependent T cell activation. H2S stimulated T cell activation by potentiating MEK-dependent ERK phosphorylation, and thrombospondin-1 inhibited this signaling in a CD47-dependent manner. Thrombospondin-1 also limited activation-dependent T cell expression of the H2S biosynthetic enzymes cystathionine β-synthase and cystathionine γ-lyase, thereby limiting the autocrine role of H2S in T cell activation. Thus, thrombospondin-1 signaling through CD47 is the first identified endogenous inhibitor of H2S signaling and constitutes a novel mechanism that negatively regulates T cell activation.
Collapse
Affiliation(s)
| | | | - Kelly Ivins-O’Keefe
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda MD, 20892
| | - David D. Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda MD, 20892
| |
Collapse
|
43
|
Crosstalk between colon cancer cells and macrophages via inflammatory mediators and CD47 promotes tumour cell migration. Eur J Cancer 2013; 49:3320-34. [PMID: 23810249 DOI: 10.1016/j.ejca.2013.06.005] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 05/08/2013] [Accepted: 06/02/2013] [Indexed: 12/17/2022]
Abstract
Tumour-associated macrophages (TAMs) of the M2 phenotype are present in the stroma of many tumours and are frequently associated with the progression of several types of cancer. We investigated the role of M2 macrophages in colon cancer progression and found that human colon cancer tissue had elevated numbers of CD68(+) (macrophage marker) cells and CD206(+) (M2 macrophage marker) cells and increased CD47 expression. To explore potential interplay between colon cancer cells and M2 macrophages, we differentiated the monocyte cell line THP-1 into M1 and M2 macrophages (CD206(high) and Th2 cytokine-secreting cells), respectively. M2 macrophages migrated faster than M1 macrophages towards SW480-conditioned medium. Similarly, M2 macrophage-conditioned medium induced SW480 cell migration and CD47 expression. Factors released by macrophages were involved in this induction. In addition, SW480 cells migrated faster when co-cultured with M2 macrophages. Inhibition of CD47 with blocking antibodies or siRNA significantly reduced the migration of SW480 cells in the presence of M2 macrophages. This effect was further decreased via blocking antibodies against the CD47 ligand signal-regulatory protein α (SIRPα). Additionally, cancer cells also secreted significant levels of IL-10, thereby promoting M2 macrophage differentiation. These findings indicate that a TAM-enriched tumour microenvironment promotes colon cancer cell migration and metastasis.
Collapse
|
44
|
Baba N, Van VQ, Wakahara K, Rubio M, Fortin G, Panzini B, Soucy G, Wassef R, Richard C, Tamaz R, Lahaie R, Bernard EJ, Caussignac Y, Leduc R, Lougnarath R, Bergeron C, Racicot MA, Bergeron F, Panzini MA, Demetter P, Franchimont D, Schäkel K, Weckbecker G, Kolbinger F, Heusser C, Huber T, Welzenbach K, Sarfati M. CD47 fusion protein targets CD172a+ cells in Crohn's disease and dampens the production of IL-1β and TNF. ACTA ACUST UNITED AC 2013; 210:1251-63. [PMID: 23669395 PMCID: PMC3674701 DOI: 10.1084/jem.20122037] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
CD172a+ cells producing IL-1β and TNF are increased in inflamed tissues in Crohn’s disease and can be targeted by CD47 fusion protein. In mice, the transfer of CD172a+ (SIRP-α) dendritic cells (DCs) elicits T cell–driven colitis, whereas treatment with CD47-Fc protein, a CD172a-binding agent, confers protection. The aim of this study was to elucidate the nature and functional properties of human CD172a+ DCs in chronic intestinal inflammation. Here, we show that CD172a+CD11c+ cells accumulate in the mesenteric lymph nodes (mLNs) and inflamed intestinal mucosa in patients with Crohn’s disease (CD). These cells are distinct from resident DCs and may coexpress markers typically associated with monocyte-derived inflammatory DCs such as CD14 and/or DC-SIGN, E-Cadherin, and/or CX3CR1. Spontaneous IL-1β and TNF production by HLA-DR+ cells in CD tissues is restricted to those expressing CD172a. An avidity-improved CD47 fusion protein (CD47-Var1) suppresses the release of a wide array of inflammatory cytokines by CD172a+ cells, which may include HLA-DR−CD172a+ neutrophils, in inflamed colonic explant cultures and impairs the ability of HLA-DR+CD172a+ cells to activate memory Th17 but not Th1 responses in mLNs. In conclusion, targeting CD172a+ cells may represent novel therapeutic perspectives for patients with CD.
Collapse
Affiliation(s)
- Nobuyasu Baba
- Immunoregulation Laboratory, The Research Center of the Centre Hospitalier de L'Université de Montréal (CRCHUM), Montreal, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Hancock DG, Guy TV, Shklovskaya E, Fazekas de St Groth B. Experimental models to investigate the function of dendritic cell subsets: challenges and implications. Clin Exp Immunol 2013; 171:147-54. [PMID: 23286941 DOI: 10.1111/cei.12027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2012] [Indexed: 11/29/2022] Open
Abstract
The dendritic cell (DC) lineage is remarkably heterogeneous. It has been postulated that specialized DC subsets have evolved in order to select and support the multitude of possible T cell differentiation pathways. However, defining the function of individual DC subsets has proven remarkably difficult, and DC subset control of key T cell fates such as tolerance, T helper cell commitment and regulatory T cell induction is still not well understood. While the difficulty in assigning unique functions to particular DC subsets may be due to sharing of functions, it may also reflect a lack of appropriate physiological in-vivo models for studying DC function. In this paper we review the limitations associated with many of the current DC models and highlight some of the underlying difficulties involved in studying the function of murine DC subsets.
Collapse
Affiliation(s)
- D G Hancock
- Centenary Institute of Cancer Medicine and Cell Biology and the Discipline of Dermatology, University of Sydney, Sydney, NSW, Australia
| | | | | | | |
Collapse
|
46
|
Intestinal dendritic cells: their role in intestinal inflammation, manipulation by the gut microbiota and differences between mice and men. Immunol Lett 2013; 150:30-40. [PMID: 23352670 DOI: 10.1016/j.imlet.2013.01.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 01/08/2013] [Accepted: 01/08/2013] [Indexed: 02/06/2023]
Abstract
The intestinal immune system maintains a delicate balance between immunogenicity against invading pathogens and tolerance of the commensal microbiota and food antigens. Dendritic cells (DC) generate primary T-cell responses, and determine whether these responses are immunogenic or tolerogenic. The regulatory role of DC is of particular importance in the gut due to the high antigenic load. Intestinal DC act as sentinels, sampling potentially pathogenic antigens but also harmless antigens including the commensal microbiota. Following antigen acquisition, intestinal DC migrate to secondary lymphoid organs to activate naive T-cells. DC also imprint specific homing properties on T-cells that they stimulate; gut DC specifically induce gut-homing properties on T-cells upon activation, enabling T-cell migration back to intestinal sites. Data regarding properties on gut DC in humans is scarce, although evidence now supports the role of DC as important players in intestinal immunity in humans. Here, we review the role of intestinal DC in shaping mucosal immune responses and directing tissue-specific T-cell responses, with a special focus on the importance of distinguishing DC subsets from macrophages at intestinal sites. We compare and contrast human DC with their murine counterparts, and discuss the ability of the gut microbiota to shape intestinal DC function, and how this may be dysregulated in inflammatory bowel disease (IBD). Lastly, we describe recent advances in the study of probiotics on intestinal DC function, including the use of soluble secreted bacterial products.
Collapse
|
47
|
Soto-Pantoja DR, Stein EV, Rogers NM, Sharifi-Sanjani M, Isenberg JS, Roberts DD. Therapeutic opportunities for targeting the ubiquitous cell surface receptor CD47. Expert Opin Ther Targets 2013; 17:89-103. [PMID: 23101472 PMCID: PMC3564224 DOI: 10.1517/14728222.2013.733699] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION CD47 is a ubiquitously expressed cell surface receptor that serves as a counter-receptor for SIRPα in recognition of self by the innate immune system. Independently, CD47 also functions as an important signaling receptor for regulating cell responses to stress. AREAS COVERED We review the expression, molecular interactions, and pathophysiological functions of CD47 in the cardiovascular and immune systems. CD47 was first identified as a potential tumor marker, and we examine recent evidence that its dysregulation contributes to cancer progression and evasion of anti-tumor immunity. We further discuss therapeutic strategies for enhancing or inhibiting CD47 signaling and applications of such agents in preclinical models of ischemia and ischemia/reperfusion injuries, organ transplantation, pulmonary hypertension, radioprotection, and cancer. EXPERT OPINION Ongoing studies are revealing a central role of CD47 for conveying signals from the extracellular microenvironment that limit cell and tissue survival upon exposure to various types of stress. Based on this key function, therapeutics targeting CD47 or its ligands thrombospondin-1 and SIRPα could have broad applications spanning reconstructive surgery, engineering of tissues and biocompatible surfaces, vascular diseases, diabetes, organ transplantation, radiation injuries, inflammatory diseases, and cancer.
Collapse
Affiliation(s)
- David R. Soto-Pantoja
- Cancer Research Training Award Fellow, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1500
| | - Erica V. Stein
- Predoctoral Cancer Research Training Award Fellow, Laboratoryof Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1500 and Microbiology and Immunology Program of the Institute for Biomedical Sciences, Departments of Microbiology, Immunology and Tropical Medicine, George Washington University, 2300 Eye St., N.W., Ross Hall, Washington, D.C. 20037
| | - Natasha M. Rogers
- Visiting Research Fellow, Division of Pulmonary Allergy and Critical Care Medicine, Vascular Medicine Institute, University of Pittsburgh School of Medicine, E1240 Biomedical Science Tower, Room E1200, 200 Lothrop Street, Pittsburgh, PA 15261
| | - Maryam Sharifi-Sanjani
- Post-doctoral Fellow, Division of Pulmonary Allergy and Critical Care Medicine, Vascular Medicine Institute, University of Pittsburgh School of Medicine, E1240 Biomedical Science Tower, Room E1200, 200 Lothrop Street, Pittsburgh, PA 15261
| | - Jeffrey S. Isenberg
- Associate Professor of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Vascular Medicine Institute, University of Pittsburgh School of Medicine, E1240 Biomedical Science Tower, Room E1258, 200 Lothrop Street, Pittsburgh, PA 15261
| | - David D. Roberts
- Chief, Biochemical Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 Room 2A33, Bethesda, MD 20892-1500
| |
Collapse
|
48
|
Bian Z, Guo Y, Luo Y, Tremblay A, Zhang X, Dharma S, Mishra A, Liu Y. CD47 deficiency does not impede polymorphonuclear neutrophil transmigration but attenuates granulopoiesis at the postacute stage of colitis. THE JOURNAL OF IMMUNOLOGY 2012. [PMID: 23203922 DOI: 10.4049/jimmunol.1201963] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Previous studies have suggested that CD47, an essential cell-surface protein, plays an important role in polymorphonuclear neutrophil (PMN) transmigration across tissue cells and extracellular matrix. In the current study, the role of CD47 in PMN transmigration and infiltration into tissues was further evaluated by investigating the function of CD47(-/-) PMN and inflammatory conditions induced in CD47(-/-) mice. Using in vitro time-course assays, we found that CD47(-/-) PMN exhibited no impediment, but slightly enhanced response to and transmigration toward, the chemoattractant fMLF. In vivo analysis in CD47(-/-) mice by inducing acute peritonitis and aggressive colitis observed consistent results, indicating that both PMN and monocytes effectively infiltrated inflammatory sites despite the absence of CD47 on these leukocytes or the surrounding tissue cells. Although PMN transmigration was not delayed in CD47(-/-) mice, fewer PMN were found in the intestine at the postacute/chronic stage of chronic colitis induced with sustained low-dose dextran sulfate sodium. Further analysis suggested that the paucity of PMN accumulation was attributable to attenuated granulopoiesis secondary to assessed lower levels of IL-17. Administration of exogenous IL-17A markedly increased PMN availability and rapidly rendered severe colitis in CD47(-/-) mice under dextran sulfate sodium treatment.
Collapse
Affiliation(s)
- Zhen Bian
- Program of Cellular Biology and Immunology, Department of Biology, Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA 30303, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Azcutia V, Stefanidakis M, Tsuboi N, Mayadas T, Croce KJ, Fukuda D, Aikawa M, Newton G, Luscinskas FW. Endothelial CD47 promotes vascular endothelial-cadherin tyrosine phosphorylation and participates in T cell recruitment at sites of inflammation in vivo. THE JOURNAL OF IMMUNOLOGY 2012; 189:2553-62. [PMID: 22815286 DOI: 10.4049/jimmunol.1103606] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
At sites of inflammation, endothelial adhesion molecules bind leukocytes and transmit signals required for transendothelial migration (TEM). We previously reported that adhesive interactions between endothelial cell CD47 and leukocyte signal regulatory protein γ (SIRPγ) regulate human T cell TEM. The role of endothelial CD47 in T cell TEM in vivo, however, has not been explored. In this study, CD47⁻/⁻ mice showed reduced recruitment of blood T cells as well as neutrophils and monocytes in a dermal air pouch model of TNF-α-induced inflammation. Reconstitution of CD47⁻/⁻ mice with wild-type bone marrow cells did not restore leukocyte recruitment to the air pouch, indicating a role for endothelial CD47. The defect in leukocyte TEM in the CD47⁻/⁻ endothelium was corroborated by intravital microscopy of inflamed cremaster muscle microcirculation in bone marrow chimera mice. In an in vitro human system, CD47 on both HUVEC and T cells was required for TEM. Although previous studies showed CD47-dependent signaling required G(αi)-coupled pathways, this was not the case for endothelial CD47 because pertussis toxin, which inactivates G(αi), had no inhibitory effect, whereas G(αi) was required by the T cell for TEM. We next investigated the endothelial CD47-dependent signaling events that accompany leukocyte TEM. Ab-induced cross-linking of CD47 revealed robust actin cytoskeleton reorganization and Src- and Pyk-2-kinase dependent tyrosine phosphorylation of the vascular endothelial-cadherin cytoplasmic tail. This signaling was pertussis toxin insensitive, suggesting that endothelial CD47 signaling is independent of G(αi). These findings suggest that engagement of endothelial CD47 by its ligands triggers outside-in signals in endothelium that facilitate leukocyte TEM.
Collapse
Affiliation(s)
- Veronica Azcutia
- Department of Pathology, Center for Excellence in Vascular Biology, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Inman CF, Laycock GM, Mitchard L, Harley R, Warwick J, Burt R, van Diemen PM, Stevens M, Bailey M. Neonatal colonisation expands a specific intestinal antigen-presenting cell subset prior to CD4 T-cell expansion, without altering T-cell repertoire. PLoS One 2012; 7:e33707. [PMID: 22442714 PMCID: PMC3307746 DOI: 10.1371/journal.pone.0033707] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 02/15/2012] [Indexed: 11/18/2022] Open
Abstract
Interactions between the early-life colonising intestinal microbiota and the developing immune system are critical in determining the nature of immune responses in later life. Studies in neonatal animals in which this interaction can be examined are central to understanding the mechanisms by which the microbiota impacts on immune development and to developing therapies based on manipulation of the microbiome. The inbred piglet model represents a system that is comparable to human neonates and allows for control of the impact of maternal factors. Here we show that colonisation with a defined microbiota produces expansion of mucosal plasma cells and of T-lymphocytes without altering the repertoire of alpha beta T-cells in the intestine. Importantly, this is preceded by microbially-induced expansion of a signal regulatory protein α-positive (SIRPα+) antigen-presenting cell subset, whilst SIRPα−CD11R1+ antigen-presenting cells (APCs) are unaffected by colonisation. The central role of intestinal APCs in the induction and maintenance of mucosal immunity implicates SIRPα+ antigen-presenting cells as orchestrators of early-life mucosal immune development.
Collapse
Affiliation(s)
- Charlotte F Inman
- School of Clinical Veterinary Science, University of Bristol, Langford, Bristol, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|