1
|
Yeyeodu S, Hanafi D, Webb K, Laurie NA, Kimbro KS. Population-enriched innate immune variants may identify candidate gene targets at the intersection of cancer and cardio-metabolic disease. Front Endocrinol (Lausanne) 2024; 14:1286979. [PMID: 38577257 PMCID: PMC10991756 DOI: 10.3389/fendo.2023.1286979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/07/2023] [Indexed: 04/06/2024] Open
Abstract
Both cancer and cardio-metabolic disease disparities exist among specific populations in the US. For example, African Americans experience the highest rates of breast and prostate cancer mortality and the highest incidence of obesity. Native and Hispanic Americans experience the highest rates of liver cancer mortality. At the same time, Pacific Islanders have the highest death rate attributed to type 2 diabetes (T2D), and Asian Americans experience the highest incidence of non-alcoholic fatty liver disease (NAFLD) and cancers induced by infectious agents. Notably, the pathologic progression of both cancer and cardio-metabolic diseases involves innate immunity and mechanisms of inflammation. Innate immunity in individuals is established through genetic inheritance and external stimuli to respond to environmental threats and stresses such as pathogen exposure. Further, individual genomes contain characteristic genetic markers associated with one or more geographic ancestries (ethnic groups), including protective innate immune genetic programming optimized for survival in their corresponding ancestral environment(s). This perspective explores evidence related to our working hypothesis that genetic variations in innate immune genes, particularly those that are commonly found but unevenly distributed between populations, are associated with disparities between populations in both cancer and cardio-metabolic diseases. Identifying conventional and unconventional innate immune genes that fit this profile may provide critical insights into the underlying mechanisms that connect these two families of complex diseases and offer novel targets for precision-based treatment of cancer and/or cardio-metabolic disease.
Collapse
Affiliation(s)
- Susan Yeyeodu
- Julius L Chambers Biomedical/Biotechnology Institute (JLC-BBRI), North Carolina Central University, Durham, NC, United States
- Charles River Discovery Services, Morrisville, NC, United States
| | - Donia Hanafi
- Julius L Chambers Biomedical/Biotechnology Institute (JLC-BBRI), North Carolina Central University, Durham, NC, United States
| | - Kenisha Webb
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Nikia A. Laurie
- Julius L Chambers Biomedical/Biotechnology Institute (JLC-BBRI), North Carolina Central University, Durham, NC, United States
| | - K. Sean Kimbro
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| |
Collapse
|
2
|
Redondo-García S, Barritt C, Papagregoriou C, Yeboah M, Frendeus B, Cragg MS, Roghanian A. Human leukocyte immunoglobulin-like receptors in health and disease. Front Immunol 2023; 14:1282874. [PMID: 38022598 PMCID: PMC10679719 DOI: 10.3389/fimmu.2023.1282874] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 09/20/2023] [Indexed: 12/01/2023] Open
Abstract
Human leukocyte immunoglobulin (Ig)-like receptors (LILR) are a family of 11 innate immunomodulatory receptors, primarily expressed on lymphoid and myeloid cells. LILRs are either activating (LILRA) or inhibitory (LILRB) depending on their associated signalling domains (D). With the exception of the soluble LILRA3, LILRAs mediate immune activation, while LILRB1-5 primarily inhibit immune responses and mediate tolerance. Abnormal expression and function of LILRs is associated with a range of pathologies, including immune insufficiency (infection and malignancy) and overt immune responses (autoimmunity and alloresponses), suggesting LILRs may be excellent candidates for targeted immunotherapies. This review will discuss the biology and clinical relevance of this extensive family of immune receptors and will summarise the recent developments in targeting LILRs in disease settings, such as cancer, with an update on the clinical trials investigating the therapeutic targeting of these receptors.
Collapse
Affiliation(s)
- Silvia Redondo-García
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Christopher Barritt
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
- Lister Department of General Surgery, Glasgow Royal Infirmary, Glasgow, United Kingdom
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, United Kingdom
| | - Charys Papagregoriou
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Muchaala Yeboah
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Björn Frendeus
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
- BioInvent International AB, Lund, Sweden
| | - Mark S. Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Ali Roghanian
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
3
|
Chan C, Lustig M, Baumann N, Valerius T, van Tetering G, Leusen JHW. Targeting Myeloid Checkpoint Molecules in Combination With Antibody Therapy: A Novel Anti-Cancer Strategy With IgA Antibodies? Front Immunol 2022; 13:932155. [PMID: 35865547 PMCID: PMC9295600 DOI: 10.3389/fimmu.2022.932155] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Immunotherapy with therapeutic antibodies has shown a lack of durable responses in some patients due to resistance mechanisms. Checkpoint molecules expressed by tumor cells have a deleterious impact on clinical responses to therapeutic antibodies. Myeloid checkpoints, which negatively regulate macrophage and neutrophil anti-tumor responses, are a novel type of checkpoint molecule. Myeloid checkpoint inhibition is currently being studied in combination with IgG-based immunotherapy. In contrast, the combination with IgA-based treatment has received minimal attention. IgA antibodies have been demonstrated to more effectively attract and activate neutrophils than their IgG counterparts. Therefore, myeloid checkpoint inhibition could be an interesting addition to IgA treatment and has the potential to significantly enhance IgA therapy.
Collapse
Affiliation(s)
- Chilam Chan
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Marta Lustig
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Niklas Baumann
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Thomas Valerius
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Geert van Tetering
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jeanette H. W. Leusen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
- *Correspondence: Jeanette H. W. Leusen,
| |
Collapse
|
4
|
Abdallah F, Coindre S, Gardet M, Meurisse F, Naji A, Suganuma N, Abi-Rached L, Lambotte O, Favier B. Leukocyte Immunoglobulin-Like Receptors in Regulating the Immune Response in Infectious Diseases: A Window of Opportunity to Pathogen Persistence and a Sound Target in Therapeutics. Front Immunol 2021; 12:717998. [PMID: 34594332 PMCID: PMC8478328 DOI: 10.3389/fimmu.2021.717998] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/25/2021] [Indexed: 12/19/2022] Open
Abstract
Immunoregulatory receptors are essential for orchestrating an immune response as well as appropriate inflammation in infectious and non-communicable diseases. Among them, leukocyte immunoglobulin-like receptors (LILRs) consist of activating and inhibitory receptors that play an important role in regulating immune responses modulating the course of disease progression. On the one hand, inhibitory LILRs constitute a safe-guard system that mitigates the inflammatory response, allowing a prompt return to immune homeostasis. On the other hand, because of their unique capacity to attenuate immune responses, pathogens use inhibitory LILRs to evade immune recognition, thus facilitating their persistence within the host. Conversely, the engagement of activating LILRs triggers immune responses and the production of inflammatory mediators to fight microbes. However, their heightened activation could lead to an exacerbated immune response and persistent inflammation with major consequences on disease outcome and autoimmune disorders. Here, we review the genetic organisation, structure and ligands of LILRs as well as their role in regulating the immune response and inflammation. We also discuss the LILR-based strategies that pathogens use to evade immune responses. A better understanding of the contribution of LILRs to host-pathogen interactions is essential to define appropriate treatments to counteract the severity and/or persistence of pathogens in acute and chronic infectious diseases lacking efficient treatments.
Collapse
Affiliation(s)
- Florence Abdallah
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Sixtine Coindre
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Margaux Gardet
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Florian Meurisse
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Abderrahim Naji
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School, Kochi University, Nankoku-City, Japan
| | - Narufumi Suganuma
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School, Kochi University, Nankoku-City, Japan
| | - Laurent Abi-Rached
- Aix-Marseille University, IRD, APHM, MEPHI, IHU Mediterranean Infection, SNC5039 CNRS, Marseille, France.,SNC5039 CNRS, Marseille, France
| | - Olivier Lambotte
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France.,Public-Hospital Assistance of Paris, Department of Internal Medicine and Clinical Immunology, Paris-Saclay University Hospital Group, Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Benoit Favier
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| |
Collapse
|
5
|
Arthur L, Esaulova E, Mogilenko DA, Tsurinov P, Burdess S, Laha A, Presti R, Goetz B, Watson MA, Goss CW, Gurnett CA, Mudd PA, Beers C, O'Halloran JA, Artyomov MN. Cellular and plasma proteomic determinants of COVID-19 and non-COVID-19 pulmonary diseases relative to healthy aging. NATURE AGING 2021; 1:535-549. [PMID: 37117829 DOI: 10.1038/s43587-021-00067-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 04/14/2021] [Indexed: 04/30/2023]
Abstract
We examine the cellular and soluble determinants of coronavirus disease 2019 (COVID-19) relative to aging by performing mass cytometry in parallel with clinical blood testing and plasma proteomic profiling of ~4,700 proteins from 71 individuals with pulmonary disease and 148 healthy donors (25-80 years old). Distinct cell populations were associated with age (GZMK+CD8+ T cells and CD25low CD4+ T cells) and with COVID-19 (TBET-EOMES- CD4+ T cells, HLA-DR+CD38+ CD8+ T cells and CD27+CD38+ B cells). A unique population of TBET+EOMES+ CD4+ T cells was associated with individuals with COVID-19 who experienced moderate, rather than severe or lethal, disease. Disease severity correlated with blood creatinine and urea nitrogen levels. Proteomics revealed a major impact of age on the disease-associated plasma signatures and highlighted the divergent contribution of hepatocyte and muscle secretomes to COVID-19 plasma proteins. Aging plasma was enriched in matrisome proteins and heart/aorta smooth muscle cell-specific proteins. These findings reveal age-specific and disease-specific changes associated with COVID-19, and potential soluble mediators of the physiological impact of COVID-19.
Collapse
Affiliation(s)
- Laura Arthur
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ekaterina Esaulova
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Denis A Mogilenko
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Petr Tsurinov
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
- JetBrains Research, Saint Petersburg, Russia
| | - Samantha Burdess
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Anwesha Laha
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Rachel Presti
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Brian Goetz
- Siteman Cancer Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Mark A Watson
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Charles W Goss
- Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Christina A Gurnett
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Philip A Mudd
- Department of Emergency Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Courtney Beers
- Siteman Cancer Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jane A O'Halloran
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
6
|
Wang M, Liu M, Jia J, Shi H, Teng J, Liu H, Sun Y, Cheng X, Ye J, Su Y, Chi H, Liu T, Wang Z, Wan L, Meng J, Ma Y, Yang C, Hu Q. Association of the Leukocyte Immunoglobulin-like Receptor A3 Gene With Neutrophil Activation and Disease Susceptibility in Adult-Onset Still's Disease. Arthritis Rheumatol 2021; 73:1033-1043. [PMID: 33381895 PMCID: PMC8252061 DOI: 10.1002/art.41635] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 12/23/2020] [Indexed: 12/21/2022]
Abstract
Objective Adult‐onset Still’s disease (AOSD) is a severe autoinflammatory disease. Neutrophil activation with enhanced neutrophil extracellular trap (NET) formation is involved in the pathogenesis of AOSD. Functional leukocyte immunoglobulin‐like receptor A3 (LIR‐A3; gene name LILRA3) has been reported to be associated with many autoimmune diseases. We aimed to investigate the association of LILRA3 with disease susceptibility and neutrophil activation in AOSD. Methods The LILRA3 deletion polymorphism and its tagging single‐nucleotide polymorphism rs103294 were genotyped in 164 patients with AOSD and 305 healthy controls. The impact of LILRA3 on clinical features and messenger RNA expression was evaluated. Plasma levels of LIR‐A3 were detected using enzyme‐linked immunosorbent assay (ELISA), and the correlation between LIR‐A3 plasma levels and disease activity and levels of circulating NET‐DNA was investigated. LIR‐A3–induced NETs were determined using PicoGreen double‐stranded DNA dye and immunofluorescence analysis in human neutrophils and a neutrophil‐like differentiated NB4 cell line transfected with LIR‐B2 small interfering RNA. Results The findings from genotyping demonstrated that functional LILRA3 was a risk factor for AOSD (11% in AOSD patients versus 5.6% in healthy controls; odds ratio 2.089 [95% confidence interval 1.030–4.291], P = 0.034), and associated with leukocytosis (P = 0.039) and increased levels of circulating neutrophils (P = 0.027). Functional LILRA3 messenger RNA expression was higher in the peripheral blood mononuclear cells (P < 0.0001) and neutrophils (P < 0.001) of LILRA3+/+ patients. Plasma levels of LIR‐A3 were elevated in patients with AOSD (P < 0.0001) and correlated with disease activity indicators and levels of circulating NET–DNA complexes. Finally, enhanced NET formation was identified in neutrophils from healthy controls and patients with inactive AOSD after stimulation of the neutrophils with LIR‐A3. Moreover, NET formation was impaired in NB4 cells after knockdown of LILRB2 gene expression. Conclusion Our study provides the first evidence that functional LILRA3 is a novel genetic risk factor for the development of AOSD and that functional LIR‐A3 may play a pathogenic role by inducing formation of NETs.
Collapse
Affiliation(s)
- Mengyan Wang
- Ruijin Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengru Liu
- Ruijin Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinchao Jia
- Ruijin Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Shi
- Ruijin Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jialin Teng
- Ruijin Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Honglei Liu
- Ruijin Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Sun
- Ruijin Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaobing Cheng
- Ruijin Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junna Ye
- Ruijin Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yutong Su
- Ruijin Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huihui Chi
- Ruijin Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Liu
- Ruijin Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhihong Wang
- Ruijin Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liyan Wan
- Ruijin Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianfen Meng
- Ruijin Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuning Ma
- Ruijin Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengde Yang
- Ruijin Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiongyi Hu
- Ruijin Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Hirayasu K, Sun J, Hasegawa G, Hashikawa Y, Hosomichi K, Tajima A, Tokunaga K, Ohashi J, Hanayama R. Characterization of LILRB3 and LILRA6 allelic variants in the Japanese population. J Hum Genet 2021; 66:739-748. [PMID: 33526815 DOI: 10.1038/s10038-021-00906-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/16/2021] [Accepted: 01/17/2021] [Indexed: 11/09/2022]
Abstract
Leukocyte immunoglobulin (Ig)-like receptors (LILRs) are encoded by members of a human multigene family, comprising 11 protein-coding genes and two pseudogenes. Among the LILRs, LILRB3 and LILRA6 show the highest homology with each other, along with high allelic and copy number variations. Therefore, it has been difficult to discriminate between them, both genetically and functionally, precluding disease association studies of LILRB3 and LILRA6. In this study, we carefully performed variant screening of LILRB3 and LILRA6 by cDNA cloning from Japanese individuals and identified four allelic lineages showing significantly high non-synonymous-to-synonymous ratios in pairwise comparisons. Furthermore, the extracellular domains of the LILRB3*JP6 and LILRA6*JP1 alleles were identical at the DNA level, suggesting that gene conversion-like events diversified LILRB3 and LILRA6. To determine the four allelic lineages from genomic DNA, we established a lineage typing method that accurately estimated the four allelic lineages in addition to specific common alleles from genomic DNA. Analysis of LILRA6 copy number variation revealed one, two, and three copies of LILRA6 in the Japanese-in-Tokyo (JPT) population. Flow cytometric analysis showed that an anti-LILRB3 antibody did not recognize the second most common lineage in the Japanese population, indicating significant amino acid differences across the allelic lineages. Taken together, our findings indicate that our lineage typing is useful for classifying the lineage-specific functions of LILRB3 and LILRA6, serving as the basis for disease association studies.
Collapse
Affiliation(s)
- Kouyuki Hirayasu
- Advanced Preventive Medical Sciences Research Center, Kanazawa University, Ishikawa, Japan. .,Department of Immunology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan.
| | - Jinwen Sun
- Department of Immunology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Gen Hasegawa
- Department of Immunology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Yuko Hashikawa
- Advanced Preventive Medical Sciences Research Center, Kanazawa University, Ishikawa, Japan
| | - Kazuyoshi Hosomichi
- Advanced Preventive Medical Sciences Research Center, Kanazawa University, Ishikawa, Japan.,Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Atsushi Tajima
- Advanced Preventive Medical Sciences Research Center, Kanazawa University, Ishikawa, Japan.,Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Katsushi Tokunaga
- Genome Medical Science Project, National Center for Global Health and Medicine, Tokyo, Japan
| | - Jun Ohashi
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Rikinari Hanayama
- Advanced Preventive Medical Sciences Research Center, Kanazawa University, Ishikawa, Japan.,Department of Immunology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan.,WPI Nano Life Science Institute (NanoLSI), Kanazawa University, Kakuma, Kanazawa, Ishikawa, Japan
| |
Collapse
|
8
|
Lan X, Liu F, Ma J, Chang Y, Lan X, Xiang L, Shen X, Zhou F, Zhao Q. Leukocyte immunoglobulin-like receptor A3 is increased in IBD patients and functions as an anti-inflammatory modulator. Clin Exp Immunol 2020; 203:286-303. [PMID: 33006756 PMCID: PMC7806419 DOI: 10.1111/cei.13529] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 12/19/2022] Open
Abstract
Growing evidence shows that a homozygous 6·7-kb deletion of the novel anti-inflammatory molecule leukocyte immunoglobulin-like receptor A3 (LILRA3) is associated with many autoimmune disorders. However, its effects on pathogenesis of inflammatory bowel disease (IBD) have yet not been clarified. LILRA3 is mainly expressed in monocytes, whereas its effects on biological behaviors of monocytes have not been systematically reported. In our study, to investigate the association between LILRA3 polymorphism and IBD susceptibility, LILRA3 polymorphism was assessed in 378 IBD patients and 509 healthy controls. Quantitative real time PCR (qRT-PCR), Western blot and immunohistochemistry (IHC) were employed to detect the LILRA3 expression in IBD patient blood and intestinal samples. The human U937 monocyte cell line was employed to establish LILRA3 over-expressing cells and the effects of LILRA3 on the biological behaviors of U937 cells were systematically explored. Although no association of the polymorphism with IBD development was found, LILRA3 expression was markedly increased in IBD patients compared with healthy controls. Over-expression of LILRA3 in monocytes led to significant decreases in secretion of interferon (IFN)-γ, tumor necrosis factor (TNF)-α and interleukin (IL)-6. Additionally, LILRA3 abated monocyte migration by reducing the expression of several chemokines and enhanced monocyte phagocytosis by increasing CD36 expression. Furthermore, LILRA3 promoted monocyte proliferation through a combination of Akt and extracellular receptor kinase/mitogen-activated protein kinase (Erk/MEK) signaling pathways. We report for the first time, to our knowledge, that LILRA3 is related to IBD and functions as an anti-inflammatory modulator in U937 cells.
Collapse
Affiliation(s)
- X Lan
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - F Liu
- Department of Gastroenterology, Xuhui District Central Hospital, Shanghai, China
| | - J Ma
- Department of Health Related Product Evaluation, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Y Chang
- Department of Gastroenterology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - X Lan
- Pathology department, National Shanghai Center for New Drug Safety Evaluation and Research, Shanghai, China
| | - L Xiang
- Department of Infectious Disease, Xiangxi Autonomous Prefecture People's Hospital, Xiangxi, China
| | - X Shen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - F Zhou
- Department of Gastroenterology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Q Zhao
- Department of Gastroenterology, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
9
|
Truong AD, Hong Y, Nguyen HT, Nguyen CT, Chu NT, Tran HTT, Dang HV, Lillehoj HS, Hong YH. Molecular identification and characterisation of a novel chicken leukocyte immunoglobulin-like receptor A5. Br Poult Sci 2020; 62:68-80. [PMID: 32812773 DOI: 10.1080/00071668.2020.1812524] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
1. Leukocyte immunoglobulin-like receptor A5 (LILRA5) is a key molecule that regulates the immune system. However, the LILRA5 gene has not been characterised in avian species, including chickens. The present study aimed to identify and functionally characterise LILRA5 identified from two genetically disparate chicken lines, viz., Marek's disease (MD)-resistant (R) line 6.3 and MD-susceptible (S) line 7.2. 2. Multiple sequence alignment and phylogenetic analyses confirmed that the identity and similarity homologies of amino acids of LILRA5 in chicken lines 6.3 and 7.2 ranged between 93% and 93.7%, whereas those between chicken and mammals ranged between 20.9% and 43.7% and 21.1% to 43.9%, respectively. The newly cloned LILRA5 from chicken lines 6.3 and 7.2 revealed high conservation and a close relationship with other known mammalian LILRA5 proteins. 3. The results indicated that LILRA5 from chicken lines 6.3 and 7.2 was associated with phosphorylation of Src kinases and protein tyrosine phosphatase non-receptor type 11 (SHP2), which play a regulatory role in immune functions. Moreover, the results demonstrated that LILRA5 in these lines was associated with the activation of major histocompatibility complex (MHC) class I and β2-microglobulin and induced the expression of the transporter associated with antigen processing. In addition, LILRA5 in both chicken lines activated and induced Janus kinase (JAK)-signal transducer and the activator of transcription (STAT), nuclear factor kappa B (NF-κB), phosphoinositide-3-kinase (PI3K)/protein kinase B (AKT) and the extracellular signal-regulated kinase (ERK)1/2 signalling pathways; toll-like receptors; and Th1-, Th2-, and Th17- cytokines. 4. The data suggested that LILRA5 has innate immune receptors essential for macrophage immune response and provide novel insights into the regulation of immunity and immunopathology.
Collapse
Affiliation(s)
- A D Truong
- Department of Biochemistry and Immunology, National Institute of Veterinary Research , Dong Da, Hanoi, Vietnam.,Department of Animal Science and Technology, Chung-Ang University , Anseong, Republic of Korea
| | - Y Hong
- Department of Animal Science and Technology, Chung-Ang University , Anseong, Republic of Korea
| | - H T Nguyen
- Department of Biochemistry and Immunology, National Institute of Veterinary Research , Dong Da, Hanoi, Vietnam
| | - C T Nguyen
- Department of Biochemistry and Immunology, National Institute of Veterinary Research , Dong Da, Hanoi, Vietnam
| | - N T Chu
- Department of Biochemistry and Immunology, National Institute of Veterinary Research , Dong Da, Hanoi, Vietnam
| | - H T T Tran
- Department of Biochemistry and Immunology, National Institute of Veterinary Research , Dong Da, Hanoi, Vietnam
| | - H V Dang
- Department of Biochemistry and Immunology, National Institute of Veterinary Research , Dong Da, Hanoi, Vietnam
| | - H S Lillehoj
- United States Department of Agriculture, Animal Biosciences and Biotechnology Laboratory, Agricultural Research Services , Beltsville, MD, USA
| | - Y H Hong
- Department of Animal Science and Technology, Chung-Ang University , Anseong, Republic of Korea
| |
Collapse
|
10
|
Research Progress on NK Cell Receptors and Their Signaling Pathways. Mediators Inflamm 2020; 2020:6437057. [PMID: 32774149 PMCID: PMC7396059 DOI: 10.1155/2020/6437057] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 05/25/2020] [Accepted: 06/20/2020] [Indexed: 12/17/2022] Open
Abstract
Natural killer cells (NK cells) play an important role in innate immunity. NK cells recognize self and nonself depending on the balance of activating receptors and inhibitory receptors. After binding to their ligands, NK cell receptors trigger subsequent signaling conduction and then determine whether NK is activated or inhibited. Furthermore, NK cell response includes cytotoxicity and cytokine release, which is tightly related to the activation of NK cell-activating receptors and the inhibition of inhibitory receptors on the surfaces of NK cells. The expression and function of NK cell surface receptors also alter in virus infection, tumor, and autoimmune diseases and influence the occurrence and development of diseases. So, it is important to understand the mechanism of recognition between NK receptors and their ligands in pathological conditions and the signaling pathways of NK cell receptors. This review mainly summarizes the research progress on NK cell surface receptors and their signal pathways.
Collapse
|
11
|
Wang H, Wang Y, Tang Y, Ye H, Zhang X, Zhou G, Lv J, Cai Y, Li Z, Guo J, Wang Q. Frequencies of the LILRA3 6.7-kb Deletion Are Highly Differentiated Among Han Chinese Subpopulations and Involved in Ankylosing Spondylitis Predisposition. Front Genet 2019; 10:869. [PMID: 31620171 PMCID: PMC6760026 DOI: 10.3389/fgene.2019.00869] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 08/20/2019] [Indexed: 12/17/2022] Open
Abstract
Introduction: Leukocyte immunoglobulin-like receptor A3 (LILRA3) belongs to the LILR family with unique feature of a 6.7-kb deletion variation among individuals. Frequencies of the 6.7-kb deletion vary widely across populations, but so far it has not been carefully investigated among Han Chinese subpopulations. Furthermore, we previously identified the non-deleted (functional) LILRA3 as a novel genetic risk for multiple autoimmune diseases. The current study aimed to investigate (i) whether frequencies of the LILRA3 6.7-kb deletion differ within Han Chinese subpopulations and (ii) whether the functional LILRA3 is a novel genetic risk for ankylosing spondylitis (AS). Methods: The LILRA3 6.7-kb deletion was genotyped in two independent cohorts, including 1,567 subjects from Shenzhen Hospital and 2,507 subjects from People’s Hospital of Peking University. Frequencies of the 6.7-kb deletion were first investigated in combined healthy cohort according to the Chinese administrative district divisions. Association analyses were performed on whole dataset and subsets according to the geographic regions. Impact of the functional LILRA3 on AS disease activity was evaluated. Results: Frequencies of LILRA3 6.7-kb deletion were highly differentiated within Han Chinese subpopulations, being gradually decreased from Northeast (80.6%) to South (47.4%). Functional LILRA3 seemed to be a strong genetic risk in susceptibility to AS under almost all the alternative genetic models, if the study subjects were not geographically stratified. However, stratification analysis revealed that the functional LILRA3 was consistently associated with AS susceptibility mainly in Northern Han subgroup under the alternative genetic models, but not in Central and Southern Hans. Functional LILRA3 conferred an increased disease activity in AS patients (P < 0.0001 both for CRP and ESR, and P = 0.003 for BASDAI). Conclusions: The present study is the first to report that the frequencies of LILRA3 6.7-kb deletion vary among Chinese Hans across geographic regions. The functional LILRA3 is associated with AS susceptibility mainly in Northern Han, but not in Central and Southern Han subgroups. Our finding provides new evidence that LILRA3 is a common genetic risk for multiple autoimmune diseases and highlights the genetic differentiation among different ethnicities, even within the subpopulations of an ethnic group.
Collapse
Affiliation(s)
- Han Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yuxuan Wang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Yundi Tang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Hua Ye
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Xuewu Zhang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Gengmin Zhou
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jiyang Lv
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yongjiang Cai
- Health Management Center, Peking University Shenzhen Hospital, Shenzhen, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Jianping Guo
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Qingwen Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
12
|
Majorczyk E, Wiśniewski A, Zoń-Giebel A, Chlebicki A, Wiland P, Kuśnierczyk P. The effect of LILRB1 but not LILRA3 gene polymorphism in immunopathology of ankylosing spondylitis-A parallel to KIR genes. Int J Immunogenet 2019; 46:146-151. [PMID: 30892832 DOI: 10.1111/iji.12422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 02/20/2019] [Accepted: 02/22/2019] [Indexed: 12/19/2022]
Abstract
LILR and KIR receptors recognize HLA-B27 and may influence immune response in ankylosing spondylitis (AS) development. Purpose of the study was to analyse LILRB1/LILRA3 polymorphisms in AS. We observed a possible protective effect of the T allele of LILRB1 rs1061680:T>C and no association with insertion/deletion polymorphisms of LILRA3 with AS.
Collapse
Affiliation(s)
- Edyta Majorczyk
- Department of Clinical Immunology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland.,Department of Biochemistry and Physiology, Faculty of Physical Education and Physiotherapy, Institute of Physiotherapy, Opole University of Technology, Opole, Poland
| | - Andrzej Wiśniewski
- Department of Clinical Immunology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Aleksandra Zoń-Giebel
- Silesian Centre for Rheumatology, Rehabilitation and Disability Prevention, Ustroń, Poland
| | - Arkadiusz Chlebicki
- Department and Clinic of Rheumatology and Internal Medicine, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Piotr Wiland
- Department and Clinic of Rheumatology and Internal Medicine, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Piotr Kuśnierczyk
- Department of Clinical Immunology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| |
Collapse
|
13
|
Vlieg HC, Huizinga EG, Janssen BJC. Structure and flexibility of the extracellular region of the PirB receptor. J Biol Chem 2019; 294:4634-4643. [PMID: 30674550 DOI: 10.1074/jbc.ra118.004396] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 01/14/2019] [Indexed: 11/06/2022] Open
Abstract
Murine paired immunoglobulin receptor B (PirB) and its human ortholog leukocyte immunoglobulin-like receptor B2 (LILRB2) are widely expressed inhibitory receptors that interact with a diverse set of extracellular ligands and exert functions ranging from down-regulation of immune responses to inhibition of neuronal growth. However, structural information that could shed light on how PirB interacts with its ligands is lacking. Here, we report crystal structures of the PirB ectodomain; the first full ectodomain structure for a LILR family member, at 3.3-4.5 Å resolution. The structures reveal that PirB's six Ig-like domains are arranged at acute angles, similar to the structures of leukocyte immunoglobulin-like receptor (LILR) and killer-cell immunoglobulin-like receptor (KIR). We observe that this regular arrangement is followed throughout the ectodomain, resulting in an extended zigzag conformation. In two out of the five structures reported here, the repeating zigzag is broken by the first domain that can adopt two alternative orientations. Quantitative binding experiments revealed a 9 μm dissociation constant for PirB-myelin-associated glycoprotein (MAG) ectodomain interactions. Taken together, these structural findings and the observed PirB-MAG interactions are compatible with a model for intercellular signaling in which the PirB extracellular domains, which point away from the cell surface, enable interaction with ligands in trans.
Collapse
Affiliation(s)
- Hedwich C Vlieg
- From Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Eric G Huizinga
- From Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Bert J C Janssen
- From Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
14
|
Genetic determinants and an epistasis of LILRA3 and HLA-B*52 in Takayasu arteritis. Proc Natl Acad Sci U S A 2018; 115:13045-13050. [PMID: 30498034 DOI: 10.1073/pnas.1808850115] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Takayasu arteritis (TAK) is a systemic vasculitis with severe complications that affects the aorta and its large branches. HLA-B*52 is an established susceptibility locus to TAK. To date, there are still only a limited number of reports concerning non-HLA susceptibility loci to TAK. We conducted a genome-wide association study (GWAS) and a follow-up study in a total of 633 TAK cases and 5,928 controls. A total of 510,879 SNPs were genotyped, and 5,875,450 SNPs were imputed together with HLA-B*52. Functional annotation of significant loci, enhancer enrichment, and pathway analyses were conducted. We identified four unreported significant loci, namely rs2322599, rs103294, rs17133698, and rs1713450, in PTK2B, LILRA3/LILRB2, DUSP22, and KLHL33, respectively. Two additional significant loci unreported in non-European GWAS were identified, namely HSPA6/FCGR3A and chr21q.22. We found that a single variant associated with the expression of MICB, a ligand for natural killer (NK) cell receptor, could explain the entire association with the HLA-B region. Rs2322599 is strongly associated with the expression of PTK2B Rs103294 risk allele in LILRA3/LILRB2 is known to be a tagging SNP for the deletion of LILRA3, a soluble receptor of HLA class I molecules. We found a significant epistasis effect between HLA-B*52 and rs103294 (P = 1.2 × 10-3). Enhancer enrichment analysis and pathway analysis suggested the involvement of NK cells (P = 8.8 × 10-5, enhancer enrichment). In conclusion, four unreported TAK susceptibility loci and an epistasis effect between LILRA3 and HLA-B*52 were identified. HLA and non-HLA regions suggested a critical role for NK cells in TAK.
Collapse
|
15
|
Leukocyte Immunoglobulin-Like Receptors A2 and A6 are Expressed in Avian Macrophages and Modulate Cytokine Production by Activating Multiple Signaling Pathways. Int J Mol Sci 2018; 19:ijms19092710. [PMID: 30208630 PMCID: PMC6163679 DOI: 10.3390/ijms19092710] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/03/2018] [Accepted: 09/04/2018] [Indexed: 12/18/2022] Open
Abstract
The activating leukocyte immunoglobulin-like receptors (LILRAs) play an important role in innate immunity. However, most of the LILRA members have not been characterized in avian species including chickens. The present study is the first attempt at cloning, structural analysis and functional characterization of two LILRAs (LILRA2 and LILRA6) in chickens. Multiple sequence alignments and construction of a phylogenetic tree of chicken LILRA2 and LILRA6 with mammalian proteins revealed high conservation between chicken LILRA2 and LILRA6 and a close relationship between the chicken and mammalian proteins. The mRNA expression of LILRA2 and LILRA6 was high in chicken HD11 macrophages and the small intestine compared to that in several other tissues and cells tested. To examine the function of LILRA2 and LILRA6 in chicken immunity, LILRA2 and LILRA6 were transfected into HD11 cells. Our findings indicated that LILRA2 and LILRA6 are associated with the phosphorylation of Src kinases and SHP2, which play a regulatory role in immune functions. Moreover, LILRA6 associated with and activated MHC class I, β2-microglobulin and induced the expression of transporters associated with antigen processing but LILRA2 did not. Furthermore, both LILRA2 and LILRA6 activated JAK-STAT, NF-κB, PI3K/AKT and ERK1/2 MAPK signaling pathways and induced Th1-, Th2- and Th17-type cytokines and Toll-like receptors. Collectively, this study indicates that LILRA2 and LILRA6 are essential for macrophage-mediated immune responses and they have the potential to complement the innate and adaptive immune system against pathogens.
Collapse
|
16
|
Takeda K, Nakamura A. Regulation of immune and neural function via leukocyte Ig-like receptors. J Biochem 2017; 162:73-80. [PMID: 28898976 DOI: 10.1093/jb/mvx036] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/11/2017] [Indexed: 01/02/2023] Open
Abstract
Leukocyte Ig-like receptors (LILRs)/Ig-like transcripts (ILTs) are expressed on innate and adaptive immune cells and maintain immune homeostasis. LILRs consist of activating and inhibitory-type receptors that regulate adequate cellular functions. LILRs were firstly identified as MHC class I receptors, therefore expression and/or polymorphisms of LILRs are reported to associate with autoimmune disorders and transplant rejection; however, recent accumulating evidences have revealed that LILRs recognize with diverse ligands including bacteria and virus. In addition, inhibitory LILRB2 (ILT4) and murine relative paired Ig-like receptor (PIR)-B are expressed on neuron and is involved in the dysregulation of central nervous system via interaction with neuronal ligands including amyloid β-protein. In this review, we summarize recent discoveries on the functions of inhibitory MHC class I receptors, and discuss their regulatory roles in immune responses and neural functions.
Collapse
Affiliation(s)
- Kazuya Takeda
- Division of Immunology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Akira Nakamura
- Division of Immunology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| |
Collapse
|
17
|
van der Touw W, Chen HM, Pan PY, Chen SH. LILRB receptor-mediated regulation of myeloid cell maturation and function. Cancer Immunol Immunother 2017. [PMID: 28638976 DOI: 10.1007/s00262-017-2023-x] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The leukocyte immunoglobulin-like receptor (LILR) family comprises a set of paired immunomodulatory receptors expressed among human myeloid and lymphocyte cell populations. While six members of LILR subfamily A (LILRA) associate with membrane adaptors to signal via immunoreceptor tyrosine-based activating motifs (ITAM), LILR subfamily B (LILRB) members signal via multiple cytoplasmic immunoreceptor tyrosine-based inhibitory motifs (ITIM). Ligand specificity of some LILR family members has been studied in detail, but new perspective into the immunoregulatory aspects of this receptor family in human myeloid cells has been limited. LILRB receptors and the murine ortholog, paired immunoglobulin-like receptor B (PIRB), have been shown to negatively regulate maturation pathways in myeloid cells including mast cells, neutrophils, dendritic cells, as well as B cells. Our laboratory further demonstrated in mouse models that PIRB regulated functional development of myeloid-derived suppressor cell and the formation of a tumor-permissive microenvironment. Based on observations from the literature and our own studies, our laboratory is focusing on how LILRs modulate immune homeostasis of human myeloid cells and how these pathways may be targeted in disease states. Integrity of this pathway in tumor microenvironments, for example, permits a myeloid phenotype that suppresses antitumor adaptive immunity. This review presents the evidence supporting a role of LILRs as myeloid cell regulators and ongoing efforts to understand the functional immunology surrounding this family.
Collapse
Affiliation(s)
- William van der Touw
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY, 10029, USA
| | - Hui-Ming Chen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY, 10029, USA
- Immunotherapy Research Center, Houston Methodist Research institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Ping-Ying Pan
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY, 10029, USA
- Immunotherapy Research Center, Houston Methodist Research institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Shu-Hsia Chen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY, 10029, USA.
- Immunotherapy Research Center, Houston Methodist Research institute, 6670 Bertner Ave, Houston, TX, 77030, USA.
| |
Collapse
|
18
|
Patent Highlights October-November 2016. Pharm Pat Anal 2017; 6:53-60. [PMID: 28248128 DOI: 10.4155/ppa-2017-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A snapshot of noteworthy recent developments in the patent literature of relevance to pharmaceutical and medical research and development.
Collapse
|
19
|
Burshtyn DN, Morcos C. The Expanding Spectrum of Ligands for Leukocyte Ig-like Receptors. THE JOURNAL OF IMMUNOLOGY 2016; 196:947-55. [PMID: 26802060 DOI: 10.4049/jimmunol.1501937] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The human leukocyte Ig-like receptor family is part of the paired receptor system. The receptors are widely expressed by various immune cells, and new functions continue to emerge. Understanding the range of functions of the receptors is of general interest because several types of pathogens exploit the receptors and genetic diversity of the receptors has been linked to various autoimmune diseases. Class I major histocompatibility molecules were the first ligands appreciated for these receptors, but the types of ligands identified over the last several years are quite diverse, including intact pathogens, immune-modulatory proteins, and molecules normally found within the CNS. This review focuses on the types of ligands described to date, how the individual receptors bind to several distinct types of ligands, and the known functional consequences of those interactions.
Collapse
Affiliation(s)
- Deborah N Burshtyn
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada; and Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Chris Morcos
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada; and Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| |
Collapse
|
20
|
Low HZ, Ahrenstorf G, Pommerenke C, Habermann N, Schughart K, Ordóñez D, Stripecke R, Wilk E, Witte T. TLR8 regulation of LILRA3 in monocytes is abrogated in human immunodeficiency virus infection and correlates to CD4 counts and virus loads. Retrovirology 2016; 13:15. [PMID: 26969150 PMCID: PMC4788896 DOI: 10.1186/s12977-016-0248-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 02/29/2016] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND LILRA3 is an immunostimulatory molecule which can conditionally induce the proliferation of cytotoxic cells. LILRA3 has a deletion genotype which is associated with multiple immune disorders. In this study, we wanted to analyze the regulation of LILRA3 and its significance in the context of HIV infection. RESULTS We analyzed a panel of TLR agonists and found that ssRNA40, a TLR8 agonist, is a potent inducer of LILRA3 in healthy individuals. However, this regulation is much diminished in HIV. Comparison of TLR8 to TLR4 induction of LILRA3 indicated that LPS induces less LILRA3 than ssRNA40 among healthy controls, but not HIV patients. Levels of LILRA3 induction correlated to virus load and CD4 counts in untreated patients. Recombinant LILRA3 can induce a host of proinflammatory genes which include IL-6 and IL-1α, as well as alter the expression of MHC and costimulatory molecules in monocytes and B-cells. CONCLUSION Our experiments point towards a beneficial role for LILRA3 in virus infections, especially in ssRNA viruses, like HIV, that engage TLR8. However, the potentially beneficial role of LILRA3 is abrogated during a HIV infection. We believe that more work has to be done to study the role of LILRA3 in infectious diseases and that there is a potential for exploring the use of LILRA3 in the treatment of virus infections.
Collapse
Affiliation(s)
- Hui Zhi Low
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Gerrit Ahrenstorf
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Claudia Pommerenke
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Nadine Habermann
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Klaus Schughart
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany.,University of Veterinary Medicine, Hannover, Germany.,University of Tennessee Health Science Center, Memphis, TN, USA
| | - David Ordóñez
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Renata Stripecke
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Esther Wilk
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Torsten Witte
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| |
Collapse
|
21
|
Ortiz MA, Núñez C, Ordóñez D, Alvarez-Cermeño JC, Martínez-Rodriguez JE, Sánchez AJ, Arroyo R, Izquierdo G, Malhotra S, Montalban X, García-Merino A, Munteis E, Alcina A, Comabella M, Matesanz F, Villar LM, Urcelay E. Influence of the LILRA3 Deletion on Multiple Sclerosis Risk: Original Data and Meta-Analysis. PLoS One 2015; 10:e0134414. [PMID: 26274821 PMCID: PMC4537248 DOI: 10.1371/journal.pone.0134414] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/08/2015] [Indexed: 01/16/2023] Open
Abstract
Background Multiple sclerosis (MS) is a neurodegenerative, autoimmune disease of the central nervous system. Genome-wide association studies (GWAS) have identified over hundred polymorphisms with modest individual effects in MS susceptibility and they have confirmed the main individual effect of the Major Histocompatibility Complex. Additional risk loci with immunologically relevant genes were found significantly overrepresented. Nonetheless, it is accepted that most of the genetic architecture underlying susceptibility to the disease remains to be defined. Candidate association studies of the leukocyte immunoglobulin-like receptor LILRA3 gene in MS have been repeatedly reported with inconsistent results. Objectives In an attempt to shed some light on these controversial findings, a combined analysis was performed including the previously published datasets and three newly genotyped cohorts. Both wild-type and deleted LILRA3 alleles were discriminated in a single-tube PCR amplification and the resulting products were visualized by their different electrophoretic mobilities. Results and Conclusion Overall, this meta-analysis involved 3200 MS patients and 3069 matched healthy controls and it did not evidence significant association of the LILRA3 deletion [carriers of LILRA3 deletion: p = 0.25, OR (95% CI) = 1.07 (0.95–1.19)], even after stratification by gender and the HLA-DRB1*15:01 risk allele.
Collapse
Affiliation(s)
- Miguel A Ortiz
- Immunology Department, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Concepción Núñez
- Immunology Department, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - David Ordóñez
- Immunogenetics & Histocompatibility, Instituto de Investigación Sanitaria Puerta de Hierro, Majadahonda, Madrid, Spain
| | - José C Alvarez-Cermeño
- Departments of Immunology and Neurology, Multiple Sclerosis Unit, Hospital Ramon y Cajal, (IRYCIS), Madrid, Spain
| | | | - Antonio J Sánchez
- Neuroimmunology, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Rafael Arroyo
- Multiple Sclerosis Unit, Neurology Department. Hospital Clínico S. Carlos, Instituto de Investigación Sanitaria S. Carlos (IdISSC), Madrid, Spain
| | | | - Sunny Malhotra
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Xavier Montalban
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Antonio García-Merino
- Neuroimmunology, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Elvira Munteis
- Neurology, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Antonio Alcina
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas (IPBLN-CSIC), Granada, Spain
| | - Manuel Comabella
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Fuencisla Matesanz
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas (IPBLN-CSIC), Granada, Spain
| | - Luisa M Villar
- Departments of Immunology and Neurology, Multiple Sclerosis Unit, Hospital Ramon y Cajal, (IRYCIS), Madrid, Spain
| | - Elena Urcelay
- Immunology Department, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| |
Collapse
|
22
|
Zhang Z, Hatano H, Shaw J, Olde Nordkamp M, Jiang G, Li D, Kollnberger S. The Leukocyte Immunoglobulin-Like Receptor Family Member LILRB5 Binds to HLA-Class I Heavy Chains. PLoS One 2015; 10:e0129063. [PMID: 26098415 PMCID: PMC4476610 DOI: 10.1371/journal.pone.0129063] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 05/04/2015] [Indexed: 11/18/2022] Open
Abstract
Objective The leukocyte immunoglobulin-like receptor (LILR) family includes inhibitory and stimulatory members which bind to classical and non-classical HLA-class I. The ligands for many LILR including LILRB5 have not yet been identified. Methods We generated C-terminal eGFP and N-terminal FLAG-tagged fusion constructs for monitoring LILR expression. We screened for LILR binding to HLA-class I by tetramer staining of 293T cells transfected with LILRA1, A4, A5 A6 and LILRB2 and LILRB5. We also studied HLA class I tetramer binding to LILRB5 on peripheral monocyte cells. LILRB5 binding to HLA-class I heavy chains was confirmed by co-immunoprecipitation. Results HLA-B27 (B27) free heavy chain (FHC) dimer but not other HLA-class I stained LILRB5-transfected 293T cells. B27 dimer binding to LILRB5 was blocked with the class I heavy chain antibody HC10 and anti-LILRB5 antisera. B27 dimers also bound to LILRB5 on peripheral monocytes. HLA-B7 and B27 heavy chains co-immunoprecipitated with LILRB5 in transduced B and rat basophil RBL cell lines. Conclusions Our findings show that class I free heavy chains are ligands for LILRB5. The unique binding specificity of LILRB5 for HLA-class I heavy chains probably results from differences in the D1 and D2 immunoglobulin-like binding domains which are distinct from other LILR which bind to β2m-associated HLA-class I.
Collapse
Affiliation(s)
- Zhiyong Zhang
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom
| | - Hiroko Hatano
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom
| | - Jacqueline Shaw
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom
| | - Marloes Olde Nordkamp
- RDM Clinical Laboratory Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Guosheng Jiang
- Department of Hemato-Oncology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Shandong Jinan, Shandong, Peoples Republic of China
| | - Demin Li
- RDM Clinical Laboratory Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Simon Kollnberger
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
23
|
Hirayasu K, Arase H. Functional and genetic diversity of leukocyte immunoglobulin-like receptor and implication for disease associations. J Hum Genet 2015; 60:703-8. [PMID: 26040207 DOI: 10.1038/jhg.2015.64] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 05/08/2015] [Accepted: 05/10/2015] [Indexed: 01/03/2023]
Abstract
Human leukocyte immunoglobulin-like receptors (LILR) are a family of 11 functional genes encoding five activating (LILRA1, 2, 4-6), five inhibitory (LILRB1-5) and one soluble (LILRA3) form. The number of LILR genes is conserved among individuals, except for LILRA3 and LILRA6, which exhibit copy-number variations. The LILR genes are rapidly evolving and showing large interspecies differences, making it difficult to analyze the functions of LILR using an animal model. LILRs are expressed on various cells such as lymphoid and myeloid cells and the expression patterns are different from gene to gene. The LILR gene expression and polymorphisms have been reported to be associated with autoimmune and infectious diseases such as rheumatoid arthritis and cytomegalovirus infection. Although human leukocyte antigen (HLA) class I is a well-characterized ligand for some LILRs, non-HLA ligands have been increasingly identified in recent years. LILRs have diverse functions, including the regulation of inflammation, immune tolerance, cell differentiation and nervous system plasticity. This review focuses on the genetic and functional diversity of the LILR family.
Collapse
Affiliation(s)
- Kouyuki Hirayasu
- Laboratory of Immunochemistry, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Hisashi Arase
- Laboratory of Immunochemistry, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| |
Collapse
|
24
|
Transcriptional profiling of peripheral CD8+T cell responses to SIVΔnef and SIVmac251 challenge reveals a link between protective immunity and induction of systemic immunoregulatory mechanisms. Virology 2014; 468-470:581-591. [PMID: 25282469 DOI: 10.1016/j.virol.2014.09.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/19/2014] [Accepted: 09/10/2014] [Indexed: 01/17/2023]
Abstract
Immunization of macaques with attenuated simian immunodeficiency virus (SIV) with deletions in nef (SIVΔnef) is shown to elicit protective immunity to infection by pathogenic SIV, yet the mechanisms that orchestrate protection and prevent pathogenesis remains unknown. We utilized whole-genome transcriptional profiling to reveal molecular signatures of protective immunity in circulating CD8+ T cells of rhesus macaques vaccinated with SIVmac239Δnef and challenged with pathogenic SIVmac251. Our findings suggest that protective immunity to pathogenic SIV infection induced by SIVmac239∆nef is associated with balanced induction of T cell activation and immunoregulatory mechanisms and dampened activation of interferon-induced signaling pathways and cytolytic enzyme production as compared with pathogenic SIVmac251 infection of unvaccinated controls. We provide evidence that protective immunity to SIVmac251 correlates with induction of biomarkers of T cell activation, differentiation, signaling, and adhesion that were down regulated in unvaccinated controls. The study highlights potential immunomodulatory networks associated with protective immunity against the virus.
Collapse
|
25
|
Singaraja RR, Tietjen I, Hovingh GK, Franchini PL, Radomski C, Wong K, vanHeek M, Stylianou IM, Lin L, Wang L, Mitnaul L, Hubbard B, Winther M, Mattice M, Legendre A, Sherrington R, Kastelein JJ, Akinsanya K, Plump A, Hayden MR. Identification of four novel genes contributing to familial elevated plasma HDL cholesterol in humans. J Lipid Res 2014; 55:1693-701. [PMID: 24891332 PMCID: PMC4109763 DOI: 10.1194/jlr.m048710] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 06/01/2014] [Indexed: 01/15/2023] Open
Abstract
While genetic determinants strongly influence HDL cholesterol (HDLc) levels, most genetic causes underlying variation in HDLc remain unknown. We aimed to identify novel rare mutations with large effects in candidate genes contributing to extreme HDLc in humans, utilizing family-based Mendelian genetics. We performed next-generation sequencing of 456 candidate HDLc-regulating genes in 200 unrelated probands with extremely low (≤10th percentile) or high (≥90th percentile) HDLc. Probands were excluded if known mutations existed in the established HDLc-regulating genes ABCA1, APOA1, LCAT, cholesteryl ester transfer protein (CETP), endothelial lipase (LIPG), and UDP-N-acetyl-α-D-galactosamine:polypeptide N-acetylgalactosaminyltransferase 2 (GALNT2). We identified 93 novel coding or splice-site variants in 72 candidate genes. Each variant was genotyped in the proband's family. Family-based association analyses were performed for variants with sufficient power to detect significance at P < 0.05 with a total of 627 family members being assessed. Mutations in the genes glucokinase regulatory protein (GCKR), RNase L (RNASEL), leukocyte immunoglobulin-like receptor 3 (LILRA3), and dynein axonemal heavy chain 10 (DNAH10) segregated with elevated HDLc levels in families, while no mutations associated with low HDLc. Taken together, we have identified mutations in four novel genes that may play a role in regulating HDLc levels in humans.
Collapse
Affiliation(s)
- Roshni R. Singaraja
- Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
- A*STAR Institute and Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ian Tietjen
- Xenon Pharmaceuticals Inc., Burnaby, BC, Canada
| | - G. Kees Hovingh
- Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | - Linus Lin
- Merck Research Laboratories, Rahway, NJ
| | | | | | | | | | | | | | | | - John J. Kastelein
- Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | - Michael R. Hayden
- A*STAR Institute and Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Centre for Molecular Medicine and Therapeutics, and Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
26
|
Devaraju P, Witte T, Schmidt RE, Gulati R, Negi VS. Immunoglobulin-like transcripts 6 (ILT6) polymorphism influences the anti-Ro60/52 autoantibody status in south Indian SLE patients. Lupus 2014; 23:1149-55. [DOI: 10.1177/0961203314538107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Introduction Systemic lupus erythematosus (SLE) is a multisystem autoimmune disorder with complex etiology. Loss of immune tolerance against self-antigens results in activation of the immune system to produce autoantibodies, which in turn contribute to the clinical manifestations of the disease. Immune cells harbor a plethora of regulatory receptors. Immunoglobulin-like transcripts (ILTs) exhibit both immune activation and inhibitory properties. Genetic defects in genes encoding these receptors may predispose to development of autoimmune diseases secondary to loss of their function. The aim of our study was to analyze the presence or absence of the 6.7 kb segment in the ILT6 gene and its association with susceptibility to SLE and its different manifestations. Method A total of 188 SLE patients and 192 age-, sex similar-, ethnicity-matched controls were recruited. They were genotyped to test the presence or absence of the 6.7 kb segment of the ILT6 gene by polymerase chain reaction. Results The mutant allele lacking the 6.7 kb gene segment had an equal frequency in patients as well as controls (20% and 18%, respectively). The mutant allele was not associated with SLE or its clinical manifestations. However, the mutant allele was associated with the presence of anti-Ro60 ( p = 0.0005, OR 3.5, 95% CI 1.8–7.1) and anti-Ro52 ( p = 0.0027, OR 2.99, 95% CI 1.5–6.06) autoantibodies. Conclusion ILT6 deletion polymorphism does not appear to be a lupus susceptibility gene in South Indian Tamils, but may behave as a genetic modifier of autoantibody phenotype by influencing the production of anti-Ro60 and anti-Ro52 autoantibodies and thus indirectly contribute to autoimmune responses in SLE.
Collapse
Affiliation(s)
| | - T Witte
- Zentrum Innere Medizin, Klinik fur Immunologie und Rheumatologie, Medizinische Hochschule Hannover (MHH), Germany
| | - RE Schmidt
- Zentrum Innere Medizin, Klinik fur Immunologie und Rheumatologie, Medizinische Hochschule Hannover (MHH), Germany
| | - R Gulati
- Genetic Services Unit, Department of Pediatrics, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India
| | - VS Negi
- Department of Clinical Immunology
| |
Collapse
|
27
|
Bashirova AA, Apps R, Vince N, Mochalova Y, Yu XG, Carrington M. Diversity of the human LILRB3/A6 locus encoding a myeloid inhibitory and activating receptor pair. Immunogenetics 2014; 66:1-8. [PMID: 24096970 PMCID: PMC3877738 DOI: 10.1007/s00251-013-0730-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 08/10/2013] [Indexed: 12/24/2022]
Abstract
Leukocyte immunoglobulin-like receptor (LILR)B3 and LILRA6 represent a pair of inhibitory/activating receptors with identical extracellular domains and unknown ligands. LILRB3 can mediate inhibitory signaling via immunoreceptor tyrosine-based inhibition motifs in its cytoplasmic tail whereas LILRA6 can signal through association with an activating adaptor molecule, FcRγ, which bears a cytoplasmic tail with an immunoreceptor tyrosine-based activation motif. The receptors are encoded by two highly polymorphic neighboring genes within the leukocyte receptor complex on human chromosome 19. Here, we report that the two genes display similar levels of single nucleotide polymorphisms with the majority of polymorphic sites being identical. In addition, the LILRA6 gene exhibits copy number variation (CNV) whereas LILRB3 does not. A screen of healthy Caucasians indicated that 32 % of the subjects possessed more than two copies of LILRA6, whereas 4 % have only one copy of the gene per diploid genome. Analysis of mRNA expression in the major fractions of PBMCs showed that LILRA6 is primarily expressed in monocytes, similarly to LILRB3, and its expression level correlates with copy number of the gene. We suggest that the LILRA6 CNV may influence the level of the activating receptor on the cell surface, potentially affecting signaling upon LILRB3/A6 ligation.
Collapse
Affiliation(s)
- Arman A. Bashirova
- Cancer and Inflammation Program, Laboratory of Experimental Immunology, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, USA
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts 02114, USA
| | - Richard Apps
- Cancer and Inflammation Program, Laboratory of Experimental Immunology, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, USA
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts 02114, USA
| | - Nicolas Vince
- Cancer and Inflammation Program, Laboratory of Experimental Immunology, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, USA
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts 02114, USA
| | - Yelizaveta Mochalova
- Cancer and Inflammation Program, Laboratory of Experimental Immunology, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, USA
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts 02114, USA
- University of Maryland, Baltimore County, Baltimore, Maryland, 21250, USA
| | - Xu G. Yu
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts 02114, USA
| | - Mary Carrington
- Cancer and Inflammation Program, Laboratory of Experimental Immunology, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, USA
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts 02114, USA
| |
Collapse
|
28
|
Low HZ, Reuter S, Topperwien M, Dankenbrink N, Peest D, Kabalak G, Stripecke R, Schmidt RE, Matthias T, Witte T. Association of the LILRA3 deletion with B-NHL and functional characterization of the immunostimulatory molecule. PLoS One 2013; 8:e81360. [PMID: 24363809 PMCID: PMC3867304 DOI: 10.1371/journal.pone.0081360] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 10/11/2013] [Indexed: 11/19/2022] Open
Abstract
LILRA3 is the sole soluble member of the LILR family. Previous studies from our group had shown that a 6.7 kb genetic deletion of LILRA3 is associated with MS and Sjögren's syndrome. An impairment of the immune response leads to a predisposition for B-NHL, so we wanted to study whether the deletion of LILRA3 is also a risk factor for B-NHL, as well as the function of LILRA3. We discovered that the frequency of the homozygous LILRA3 deletion was significantly higher in B-NHL (6%) than in blood donors (3%) (P = 0.03). We detected binding of fluorochrome-conjugated recombinant LILRA3 to monocytes and B-cells. Incubation of PBMCs with recombinant LILRA3 induced proliferation of CD8(+) T-cells and NK cells, as determined by CFSE staining. Using a transwell system, we demonstrated that LILRA3-stimulated lymphocyte proliferation was mediated by monocytes and required both cell contact and soluble factors. Secretion of IL-6, IL-8, IL-1β and IL-10 in the cell supernatant was stimulated by LILRA3. We conclude that LILRA3 is an immunostimulatory molecule, whose deficiency is associated with higher frequency of B-NHL.
Collapse
Affiliation(s)
- Hui Zhi Low
- Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
- * E-mail:
| | | | - Michael Topperwien
- Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | - Nadine Dankenbrink
- Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | - Dietrich Peest
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Gamze Kabalak
- Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | - Renata Stripecke
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Reinhold E. Schmidt
- Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | | | - Torsten Witte
- Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
29
|
Marchesi M, Andersson E, Villabona L, Seliger B, Lundqvist A, Kiessling R, Masucci GV. HLA-dependent tumour development: a role for tumour associate macrophages? J Transl Med 2013; 11:247. [PMID: 24093459 PMCID: PMC3856519 DOI: 10.1186/1479-5876-11-247] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 10/01/2013] [Indexed: 02/06/2023] Open
Abstract
HLA abnormalities on tumour cells for immune escape have been widely described. In addition, cellular components of the tumour microenvironment, in particular myeloid derived suppressor cells (MDSC) and alternatively activated M2 tumour-associated macrophages (TAMs), are involved in tumour promotion, progression, angiogenesis and suppression of anti-tumour immunity. However, the role of HLA in these activities is poorly understood. This review details MHC class I characteristics and describes MHC class I receptors functions. This analysis established the basis for a reflection about the crosstalk among the tumour cells, the TAMs and the cells mediating an immune response.The tumour cells and TAMs exploit MHC class I molecules to modulate the surrounding immune cells. HLA A, B, C and G molecules down-regulate the macrophage myeloid activation through the interaction with the inhibitory LILRB receptors. HLA A, B, C are able to engage inhibitory KIR receptors negatively regulating the Natural Killer and cytotoxic T lymphocytes function while HLA-G induces the secretion of pro-angiogenic cytokines and chemokine thanks to an activator KIR receptor expressed by a minority of peripheral NK cells. The open conformer of classical MHC-I is able to interact with LILRA receptors described as being associated to the Th2-type cytokine response, triggering a condition for the M2 like TAM polarization. In addition, HLA-E antigens on the surface of the TAMs bind the inhibitory receptor CD94/NKG2A expressed by a subset of NK cells and activated cytotoxic T lymphocytes protecting from the cytolysis.Furthermore MHC class II expression by antigen presenting cells is finely regulated by factors provided with immunological capacities. Tumour-associated macrophages show an epigenetically controlled down-regulation of the MHC class II expression induced by the decoy receptor DcR3, a member of the TNFR, which further enhances the M2-like polarization. BAT3, a positive regulator of MHC class II expression in normal macrophages, seems to be secreted by TAMs, consequently lacking its intracellular function, it looks like acting as an immunosuppressive factor.In conclusion HLA could cover a considerable role in tumour-development orchestrated by tumour-associated macrophages.
Collapse
Affiliation(s)
- Maddalena Marchesi
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
- Roche Pharma, Basel, Switzerland
| | - Emilia Andersson
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Lisa Villabona
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle/Saale, Germany
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Rolf Kiessling
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Giuseppe V Masucci
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| |
Collapse
|
30
|
Nam G, Shi Y, Ryu M, Wang Q, Song H, Liu J, Yan J, Qi J, Gao GF. Crystal structures of the two membrane-proximal Ig-like domains (D3D4) of LILRB1/B2: alternative models for their involvement in peptide-HLA binding. Protein Cell 2013; 4:761-70. [PMID: 23955630 DOI: 10.1007/s13238-013-3908-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 07/03/2013] [Indexed: 10/26/2022] Open
Abstract
Leukocyte immunoglobulin-like receptors (LILRs), also called CD85s, ILTs, or LIRs, are important mediators of immune activation and tolerance that contain tandem immunoglobulin (Ig)-like folds. There are 11 (in addition to two pseudogenes) LILRs in total, two with two Ig-like domains (D1D2) and the remaining nine with four Ig-like domains (D1D2D3D4). Thus far, the structural features of the D1D2 domains of LILR proteins are well defined, but no structures for the D3D4 domains have been reported. This is a very important field to be studied as it relates to the unknown functions of the D3D4 domains, as well as their relative orientation to the D1D2 domains on the cell surface. Here, we report the crystal structures of the D3D4 domains of both LILRB1 and LILRB2. The two Ig-like domains of both LILRB1-D3D4 and LILRB2-D3D4 are arranged at an acute angle (∼60°) to form a bent structure, resembling the structures of natural killer inhibitory receptors. Based on these two D3D4 domain structures and previously reported D1D2/HLA I complex structures, two alternative models of full-length (four Ig-like domains) LILR molecules bound to HLA I are proposed.
Collapse
Affiliation(s)
- Gol Nam
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
LILRA3 is associated with benign prostatic hyperplasia risk in a Chinese Population. Int J Mol Sci 2013; 14:8832-40. [PMID: 23615473 PMCID: PMC3676759 DOI: 10.3390/ijms14058832] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 03/21/2013] [Accepted: 04/08/2013] [Indexed: 11/17/2022] Open
Abstract
A recent prostate cancer (PCa) genome-wide association study (GWAS) identified rs103294, a single nucleotide polymorphism (SNP) located on LILRA3, a key component in the regulation of inflammatory inhibition, to be significantly associated with PCa risk in a Chinese population. Because inflammation may be a common etiological risk factor between PCa and benign prostatic hyperplasia (BPH), the current study was conducted to investigate the association of rs103294 with BPH risk. rs103294 was genotyped in a Chinese population of 426 BPH cases and 1,008 controls from Xinhua Hospital in Shanghai, China. Association between rs103294, BPH risk and clinicopathological traits were tested with adjustment for age. rs103294 was significantly associated with BPH risk with a p-value of 0.0067. Individuals with risk allele "C" had increased risk for BPH (OR = 1.34, 95% CI: 1.09-1.66). Stratified analysis revealed a stronger association risk for younger patients who are below 72 years old (OR = 1.51, 95% CI: 1.06-2.16). Our study represents the first effort to demonstrate that LILRA3 gene is significantly associated with BPH risk in a Chinese population. Our results support a common role of inflammation in the development of PCa and BPH. Additional studies are needed to further evaluate our results.
Collapse
|
32
|
Kuroki K, Furukawa A, Maenaka K. Molecular recognition of paired receptors in the immune system. Front Microbiol 2012; 3:429. [PMID: 23293633 PMCID: PMC3533184 DOI: 10.3389/fmicb.2012.00429] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 12/06/2012] [Indexed: 12/13/2022] Open
Abstract
Cell surface receptors are responsible for regulating cellular function on the front line, the cell membrane. Interestingly, accumulating evidence clearly reveals that the members of cell surface receptor families have very similar extracellular ligand-binding regions but opposite signaling systems, either inhibitory or stimulatory. These receptors are designated as paired receptors. Paired receptors often recognize not only physiological ligands but also non-self ligands, such as viral and bacterial products, to fight infections. In this review, we introduce several representative examples of paired receptors, focusing on two major structural superfamilies, the immunoglobulin-like and the C-type lectin-like receptors, and explain how these receptors distinguish self and non-self ligands to maintain homeostasis in the immune system. We further discuss the evolutionary aspects of these receptors as well as the potential drug targets for regulating diseases.
Collapse
Affiliation(s)
- Kimiko Kuroki
- Laboratory of Biomolecular Science, Faculty of Pharmaceutical Sciences, Hokkaido University Sapporo, Japan
| | | | | |
Collapse
|
33
|
Wiśniewski A, Wagner M, Nowak I, Bilińska M, Pokryszko-Dragan A, Jasek M, Kuśnierczyk P. 6.7-kbp deletion in LILRA3 (ILT6) gene is associated with later onset of the multiple sclerosis in a Polish population. Hum Immunol 2012; 74:353-7. [PMID: 23238213 DOI: 10.1016/j.humimm.2012.12.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 11/20/2012] [Accepted: 12/03/2012] [Indexed: 11/19/2022]
Abstract
Recently published studies have implicated the deletion polymorphism in LILRA3 gene, as being associated with multiple sclerosis (MS). A total of 309 patients diagnosed with MS and 379 unrelated healthy volunteers were typed for 6.7-kbp deletion in LILRA3 gene. Simultaneously, presence or absence of HLA-DRB1(∗)1501 allele was established to assess the possibility of interaction between LILRA3 deletion and HLA-DRB1(∗)1501 status. In contrast to previous reports, we did not find any association of LILRA3 deletion with MS susceptibility. Also, the HLA-DRB1(∗)1501 stratification analysis showed no LILRA3 association with the disease. However, we observed that patients negative for the deletion may begin to suffer from MS significantly earlier than patients who are positive (p = 0.014). Similarly to the most European populations we found significantly higher frequency of HLA-DRB1(∗)1501 allele in cases than we found in controls (27.0% vs. 12.5%; p < 0.0001, OR = 2.6, 95%CI = 1.96-3.42).
Collapse
Affiliation(s)
- Andrzej Wiśniewski
- Laboratory of Immunogenetics and Tissue Immunology, Department of Clinical Immunology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland.
| | | | | | | | | | | | | |
Collapse
|
34
|
Kaur G, Trowsdale J, Fugger L. Natural killer cells and their receptors in multiple sclerosis. ACTA ACUST UNITED AC 2012; 136:2657-76. [PMID: 22734127 DOI: 10.1093/brain/aws159] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The immune system has crucial roles in the pathogenesis of multiple sclerosis. While the adaptive immune cell subsets, T and B cells, have been the main focus of immunological research in multiple sclerosis, it is now important to realize that the innate immune system also has a key involvement in regulating autoimmune responses in the central nervous system. Natural killer cells are innate lymphocytes that play vital roles in a diverse range of infections. There is evidence that they influence a number of autoimmune conditions. Recent studies in multiple sclerosis and its murine model, experimental autoimmune encephalomyelitis, are starting to provide some understanding of the role of natural killer cells in regulating inflammation in the central nervous system. Natural killer cells express a diverse range of polymorphic cell surface receptors, which interact with polymorphic ligands; this interaction controls the function and the activation status of the natural killer cell. In this review, we discuss evidence for the role of natural killer cells in multiple sclerosis and experimental autoimmune encephalomyelitis. We consider how a change in the balance of signals received by the natural killer cell influences its involvement in the ensuing immune response, in relation to multiple sclerosis.
Collapse
Affiliation(s)
- Gurman Kaur
- MRC Human Immunology Unit, Nuffield Department of Medicine, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | | | | |
Collapse
|
35
|
Lichterfeld M, Yu XG. The emerging role of leukocyte immunoglobulin-like receptors (LILRs) in HIV-1 infection. J Leukoc Biol 2011; 91:27-33. [PMID: 22028331 DOI: 10.1189/jlb.0811442] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
LILRs represent a group of immunomodulatory molecules that regulate the functional properties of professional APCs and influence immune activation in a variety of disease contexts. Many members of the LILR family recognize peptide/MHC class I complexes as their physiological ligands, and increasing evidence suggests that such interactions are prominently influenced by polymorphisms in HLA class I alleles or sequence variations in the presented antigenic peptides. Emerging data show that LILRs are involved in multiple, different aspects of HIV-1 disease pathogenesis and may critically influence spontaneous HIV-1 disease progression. Here, we review recent progress in understanding the role of LILR during HIV-1 infection by focusing on the dynamic interplay between LILR and HLA class I molecules in determining HIV-1 disease progression, the effects of HIV-1 mutational escape on LILR-mediated immune recognition, the contribution of LILR to HIV-1-associated immune dysfunction, and the unique expression patterns of LILR on circulating myeloid DCs from elite controllers, a small subset of HIV-1-infected patients with natural control of HIV-1 replication. Obtaining a more complete understanding of LILR-mediated immune regulation during HIV-1 infection may ultimately allow for improved strategies to treat or prevent HIV-1-associated disease manifestations.
Collapse
Affiliation(s)
- Mathias Lichterfeld
- Infectious Disease Division, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | |
Collapse
|