151
|
Kober DL, Brett TJ. TREM2-Ligand Interactions in Health and Disease. J Mol Biol 2017; 429:1607-1629. [PMID: 28432014 DOI: 10.1016/j.jmb.2017.04.004] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/06/2017] [Accepted: 04/10/2017] [Indexed: 01/04/2023]
Abstract
The protein triggering receptor expressed on myeloid cells-2 (TREM2) is an immunomodulatory receptor with a central role in myeloid cell activation and survival. In recent years, the importance of TREM2 has been highlighted by the identification of coding variants that increase risk for Alzheimer's disease and other neurodegenerative diseases. Animal studies have further shown the importance of TREM2 in neurodegenerative and other inflammatory disease models including chronic obstructive pulmonary disease, multiple sclerosis, and stroke. A mechanistic understanding of TREM2 function remains elusive, however, due in part to the absence of conclusive information regarding the identity of endogenous TREM2 ligands. While many TREM2 ligands have been proposed, their physiological role and mechanism of engagement remain to be determined. In this review, we highlight the suggested roles of TREM2 in these diseases and the recent advances in our understanding of TREM2 and discuss putative TREM2-ligand interactions and their potential roles in signaling during health and disease. We develop a model based on the TREM2 structure to explain how different TREM2 ligands might interact with the receptor and how disease risk variants may alter ligand interactions. Finally, we propose future experimental directions to establish the role and importance of these different interactions on TREM2 function.
Collapse
Affiliation(s)
- Daniel L Kober
- Molecular Microbiology and Microbial Pathogenesis Program, Washington University School of Medicine, St. Louis, MO 63110, USA; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tom J Brett
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
152
|
Jelenčić V, Lenartić M, Wensveen FM, Polić B. NKG2D: A versatile player in the immune system. Immunol Lett 2017; 189:48-53. [PMID: 28414183 DOI: 10.1016/j.imlet.2017.04.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 04/11/2017] [Indexed: 12/21/2022]
Abstract
NKG2D is known as a potent activating receptor of the immune system. It is expressed on a multitude of immune cells, including NK cells and different subsets of T cells. NKG2D recognizes various MHC I-like ligands that are induced on target cells exposed to stressors such as viral infection, DNA damage and oncological transformation. NKG2D drives or facilitates cytotoxic and cytokine responses towards cells expressing its ligands to eliminate the threat. Therefore, NKG2D is usually classified as a sensor that translates cellular stress into activation signals for immune cells. However, more recently it has become evident that NKG2D plays a role beyond direct killing of target cells. Lack of NKG2D affects development of NK cells in the bone marrow, resulting in hyperreactive NK cells. NKG2D deficiency on CD8 T cells affects the ability of effector cells to produce cytokines in response to T cell receptor engagement and reduces their capacity to establish immunological memory. Although NKG2D is not expressed on B cells subsets, lack of this receptor in hematopoietic precursors affects B cell development. Homing of mature B2 cells is altered in NKG2D-deficient mice and they have a strong reduction in peripheral B1a cell numbers, resulting in increased susceptibility to bacterial infections. The exact molecular mechanisms via which NKG2D mediates these versatile functions is still being explored, but appears to depend on the control of activation thresholds, either in hematopoietic precursors or mature immune cell subsets. In this review, we will elaborate on the underappreciated developmental and regulatory roles of NKG2D.
Collapse
Affiliation(s)
- Vedrana Jelenčić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Maja Lenartić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Felix M Wensveen
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia; Department of Experimental Immunology, Amsterdam Medical Center, Amsterdam, The Netherlands
| | - Bojan Polić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.
| |
Collapse
|
153
|
Jendresen C, Årskog V, Daws MR, Nilsson LNG. The Alzheimer's disease risk factors apolipoprotein E and TREM2 are linked in a receptor signaling pathway. J Neuroinflammation 2017; 14:59. [PMID: 28320424 PMCID: PMC5360024 DOI: 10.1186/s12974-017-0835-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 03/07/2017] [Indexed: 11/19/2022] Open
Abstract
Background Triggering receptor expressed on myeloid cells 2 (TREM2) and apolipoprotein E (APOE) are genetically linked to Alzheimer’s disease. Here, we investigated whether human ApoE mediates signal transduction through human and murine TREM2 and sought to identify a TREM2-binding domain in human ApoE. Methods To investigate cell signaling through TREM2, a cell line was used which expressed an NFAT-inducible β-galactosidase reporter and human or murine TREM2, fused to CD8 transmembrane and CD3ζ intracellular signaling domains. ELISA-based binding assays were used to determine binding affinities of human ApoE isoforms to human TREM2 and to identify a TREM2-binding domain in ApoE. Results ApoE was found to be an agonist to human TREM2 with EC50 in the low nM range, and to murine TREM2 with reduced potency. In the reporter cells, TREM2 expression was lower than in nontransgenic mouse brain. Human ApoE isoforms ε2, ε3, and ε4 bound to human TREM2 with Kd in the low nM range. The binding was displaced by an ApoE-mimetic peptide (amino acids 130–149). Conclusions An ApoE-mediated dose-dependent signal transduction through TREM2 in reporter cells was demonstrated, and a TREM2-binding region in ApoE was identified. The relevance of an ApoE-TREM2 receptor signaling pathway to Alzheimer’s disease is discussed. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0835-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Charlotte Jendresen
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Postboks 1057 Blindern, 0316, Oslo, Norway
| | - Vibeke Årskog
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Postboks 1057 Blindern, 0316, Oslo, Norway
| | - Michael R Daws
- Division of Anatomy, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Lars N G Nilsson
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Postboks 1057 Blindern, 0316, Oslo, Norway.
| |
Collapse
|
154
|
Yang Z, Lv Q, Wang Z, Dong X, Yang R, Zhao W. Identification of crucial genes associated with rat traumatic spinal cord injury. Mol Med Rep 2017; 15:1997-2006. [PMID: 28260098 PMCID: PMC5364992 DOI: 10.3892/mmr.2017.6267] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 12/09/2016] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to investigate the key genes associated with traumatic spinal cord injuries (TSCI). The dataset GSE52763 was downloaded from the Gene Expression Omnibus, for which lumbar spinal cord samples were obtained from rats at 1 and 3 weeks following contusive spinal cord injury and 1 week subsequent to a sham laminectomy, and used to identify differentially expressed genes (DEGs). Functional enrichment analysis, co‑expression analysis and transcription factor (TF) identification were performed for DEGs common to the 1 and 3 week injury samples. In total, 234 upregulated and 51 downregulated DEGs were common to the 1 and 3 week injury samples. The upregulated DEGs were significantly enriched in Gene Ontology terms concerning immunity (e.g. Itgal and Ccl2) and certain pathways, including natural killer cell mediated cytotoxicity [e.g. Ras‑related C3 botulinum toxin substrate 2 (Rac2) and TYRO protein tyrosine kinase binding protein (Tyrobp)]. The downregulated DEGs were highly enriched in female gonad development [e.g. progesterone receptor (Pgr)], and the steroid biosynthesis pathway. A total of 139 genes had co‑expression associations and the majority of them were upregulated genes. The upregulated co‑expressed genes were predominantly enriched in biological regulation, including TGFB induced factor homeobox 1 (Tgif1) and Rac2. The downregulated co‑expressed genes were enriched in anatomical structure development (e.g. Dnm3). A total of 92 co‑expressed genes composed the protein‑protein interaction network. Additionally, 9 TFs (e.g. Pgr and Tgif1) were identified from the DEGs. It was hypothesized that the genes including Tgif1, Rac2, Tyrobp, and Pgr may be closely associated with TSCI.
Collapse
Affiliation(s)
- Zibin Yang
- Department of Spinal Surgery, The People's Hospital of Dali Prefecture, Dali, Yunnan 671000, P.R. China
| | - Qiao Lv
- Department of Spinal Surgery, The People's Hospital of Dali Prefecture, Dali, Yunnan 671000, P.R. China
| | - Zhengxiang Wang
- Department of Spinal Surgery, The People's Hospital of Dali Prefecture, Dali, Yunnan 671000, P.R. China
| | - Xiliang Dong
- Department of Spinal Surgery, The People's Hospital of Dali Prefecture, Dali, Yunnan 671000, P.R. China
| | - Rongxin Yang
- Department of Spinal Surgery, The People's Hospital of Dali Prefecture, Dali, Yunnan 671000, P.R. China
| | - Wei Zhao
- Department of Spinal Surgery, The People's Hospital of Dali Prefecture, Dali, Yunnan 671000, P.R. China
| |
Collapse
|
155
|
Abstract
Triggering of cell-mediated immunity is largely dependent on the recognition of foreign or abnormal molecules by a myriad of cell surface-bound receptors. Many activating immune receptors do not possess any intrinsic signaling capacity but instead form noncovalent complexes with one or more dimeric signaling modules that communicate with a common set of kinases to initiate intracellular information-transfer pathways. This modular architecture, where the ligand binding and signaling functions are detached from one another, is a common theme that is widely employed throughout the innate and adaptive arms of immune systems. The evolutionary advantages of this highly adaptable platform for molecular recognition are visible in the variety of ligand-receptor interactions that can be linked to common signaling pathways, the diversification of receptor modules in response to pathogen challenges, and the amplification of cellular responses through incorporation of multiple signaling motifs. Here we provide an overview of the major classes of modular activating immune receptors and outline the current state of knowledge regarding how these receptors assemble, recognize their ligands, and ultimately trigger intracellular signal transduction pathways that activate immune cell effector functions.
Collapse
Affiliation(s)
- Richard Berry
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University , Clayton, Victoria 3800, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, Monash University , Clayton, Victoria 3800, Australia
| | - Matthew E Call
- Structural Biology Division, The Walter and Eliza Hall Institute of Medical Research , Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne , Parkville, Victoria 3052, Australia
| |
Collapse
|
156
|
Thiruchelvam-Kyle L, Hoelsbrekken SE, Saether PC, Bjørnsen EG, Pende D, Fossum S, Daws MR, Dissen E. The Activating Human NK Cell Receptor KIR2DS2 Recognizes a β 2-Microglobulin-Independent Ligand on Cancer Cells. THE JOURNAL OF IMMUNOLOGY 2017; 198:2556-2567. [PMID: 28202613 DOI: 10.4049/jimmunol.1600930] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 01/22/2017] [Indexed: 01/01/2023]
Abstract
The functions of activating members of the killer cell Ig-like receptor (KIR) family are not fully understood, as the ligands for these receptors are largely unidentified. In this study, we report that KIR2DS2 reporter cells recognize a ligand expressed by cancer cell lines. All cancer targets recognized by KIR2DS2 were also recognized by KIR2DL2 and KIR2DL3 reporters. Trogocytosis of membrane proteins from the cancer targets was observed with responding reporter cells, indicating the formation of KIR2DS2 ligand-specific immunological synapses. HLA-C typing of target cells showed that KIR2DS2 recognition was independent of the HLA C1 or C2 group, whereas targets cells that were only recognized by KIR2DL3 expressed C1 group alleles. Anti-HLA class I Abs blocked KIR2DL3 responses toward C1-expressing targets, but they did not block KIR2DS2 recognition of cancer cells. Small interfering RNA knockdown of β2-microglobulin reduced the expression of class I H chain on the cancer targets by >97%, but it did not reduce the KIR2DS2 reporter responses, indicating a β2-microglobulin-independent ligand for KIR2DS2. Importantly, KIR2DL3 responses toward some KIR2DS2 ligand-expressing cells were also undiminished after β2-microglobulin knockdown, and they were not blocked by anti-HLA class I Abs, suggesting that KIR2DL3, in addition to the traditional HLA-C ligands, can bind to the same β2-microglobulin-independent ligand as KIR2DS2. These observations indicate the existence of a novel, presently uncharacterized ligand for the activating NK cell receptor KIR2DS2. Molecular identification of this ligand may lead to improved KIR-HLA mismatching in hematopoietic stem cell transplantation therapy for leukemia and new, more specific NK cell-based cancer therapies.
Collapse
Affiliation(s)
- Lavanya Thiruchelvam-Kyle
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway; and
| | - Sigurd E Hoelsbrekken
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway; and
| | - Per C Saether
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway; and
| | - Elisabeth Gyllensten Bjørnsen
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway; and
| | - Daniela Pende
- Laboratorio Immunologia, Istituto di Ricovero e Cura a Carattere Scientifico Azienda Ospedaliera Universitaria San Martino-Istituto Nazionale per la Ricerca sul Cancro, 16132 Genoa, Italy
| | - Sigbjørn Fossum
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway; and
| | - Michael R Daws
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway; and
| | - Erik Dissen
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway; and
| |
Collapse
|
157
|
Zhang L, Rickard CM. Non-culture based diagnostics for intravascular catheter related bloodstream infections. Expert Rev Mol Diagn 2017; 17:181-188. [PMID: 28004592 DOI: 10.1080/14737159.2017.1275964] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
INTRODUCTION intravascular catheter related bloodstream infection (IVC-BSI) is a leading cause of nosocomial infections and associated with significant morbidity and mortality. Early detection and adequate treatment of causative pathogens is critical for a favourable outcome. However, it takes significant time to receive microbiological results due to the current reference diagnostic method's reliance on microbial growth. Areas covered: This review discusses culture and non-culture based techniques for the diagnosis of non IVC-BSI and IVC-BSI, including molecular methods and biomarkers. Different diagnostic strategies are evaluated and the potential of new generation of diagnostic assays highlighted. Expert commentary: The development of additional diagnostic methods has potential to beneficially supplement conventional culture diagnosis, and molecular techniques have particular potential to fulfil this need. They would also contribute significant new knowledge on the bacterial species present on catheters that are generally missed by diagnosis using traditionally culture-dependent methods. Advances in molecular strategies, together with new biomarkers, might lead to the development of faster, more sensitive and cheaper technologies and instruments. This review aims to provide a platform for the further development of IVCBSI diagnostic techniques.
Collapse
Affiliation(s)
- Li Zhang
- a Griffith University Menzies Health Institute Queensland , Alliance for Vascular Access Teaching and Research (AVATAR) , Nathan , Australia
| | - C M Rickard
- a Griffith University Menzies Health Institute Queensland , Alliance for Vascular Access Teaching and Research (AVATAR) , Nathan , Australia
| |
Collapse
|
158
|
Kannan GS, Aquino-Lopez A, Lee DA. Natural killer cells in malignant hematology: A primer for the non-immunologist. Blood Rev 2016; 31:1-10. [PMID: 27665023 DOI: 10.1016/j.blre.2016.08.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 08/03/2016] [Accepted: 08/29/2016] [Indexed: 12/19/2022]
Abstract
Natural killer cells were first described over 40years ago, but the last 15years has shown tremendous progress in our understanding of their biology and our ability to manipulate them for clinical therapeutic effect. Despite the increased understanding by clinicians and scientists investigating these cells, their biology remains a confusing subject for many because of the wide array of receptors, complex interactions, multiple models of predicting function, and contradictory data in the literature. While they are microscopically indistinguishable from T cells and share many of the same effector functions, their mechanisms of target recognition are completely distinct from yet complimentary to T cells. In this review we provide a basic understanding of NK cell biology and HLA recognition as compared and contrasted to T cells using a metaphor of border patrol and passports. We conclude with a summary of the evidence for NK cell effects in hematologic malignancies and describe new advances in NK cell immunotherapy aimed at improving these effects.
Collapse
Affiliation(s)
- Geoffrey S Kannan
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, 450 Brooklyn Ave, Boston, MA 02215, USA.
| | - Arianexys Aquino-Lopez
- Clinical and Translational Sciences Program, University of Texas Graduate School of Biomedical Sciences, 6767 Bertner Avenue, Houston, TX 77030, USA; Division of Pediatrics, MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 853, Houston, TX 77030, USA.
| | - Dean A Lee
- Division of Hematology, Oncology, and BMT, Nationwide Children's Hospital, 700 Children's Drive, WA4023, Columbus, OH 43205, USA.
| |
Collapse
|
159
|
Huang YQ, Li PY, Wang JB, Zhou HQ, Yang ZR, Yang RC, Bai ZF, Wang LF, Li JY, Liu HH, Zhao YL, Xiao XH. Inhibition of Sophocarpine on Poly I: C/D-GalN-Induced Immunological Liver Injury in Mice. Front Pharmacol 2016; 7:256. [PMID: 27570511 PMCID: PMC4981750 DOI: 10.3389/fphar.2016.00256] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 08/02/2016] [Indexed: 11/13/2022] Open
Abstract
Increasing evidence has suggested that natural killer (NK) cells contribute to the pathogenesis of human immunological liver injury (ILI). Previous studies have demonstrated that Sophocarpine exerts activity in immune modulation. It also has a therapeutic effect on liver protection in that it can alleviate liver fibrosis by suppressing both the activation of hepatic stellate cells and the proliferation of the activated hepatic stellate cells. However, whether Sophocarpine protects the liver by regulating NK cell activity remains unclear. In this study, the modulating effect of Sophocarpine on NK cells in the liver was investigated. The results showed that Sophocarpine dramatically decreased the production of pro-inflammatory cytokines and attenuated the liver injury induced by Poly I: C/D-GalN in C57BL/6- mice. More importantly, Sophocarpine pre-treatment significantly suppressed NK cell activation and downregulated the expression of NKG2D, a receptor responsible for NK cell activation. Moreover, the protein levels of DAP12, ZAP76 and Syk decreased, as did their corresponding mRNA levels. Overall, our study demonstrates that Sophocarpine inhibits NK cell activity, thus making it a promising therapy for ILI.
Collapse
Affiliation(s)
- Yin-Qiu Huang
- Pharmacy College, Chengdu University of Traditional Chinese MedicineChengdu, China
- Department of Pharmacy, 302 Military Hospital of ChinaBeijing, China
| | - Peng-Yan Li
- China Military Institute of Chinese Medicine, 302 Military Hospital of ChinaBeijing, China
| | - Jia-Bo Wang
- China Military Institute of Chinese Medicine, 302 Military Hospital of ChinaBeijing, China
| | - Hou-Qin Zhou
- Pharmacy College, Chengdu University of Traditional Chinese MedicineChengdu, China
- Department of Pharmacy, 302 Military Hospital of ChinaBeijing, China
| | - Zhi-Rui Yang
- Pharmacy College, Chengdu University of Traditional Chinese MedicineChengdu, China
- Department of Pharmacy, 302 Military Hospital of ChinaBeijing, China
| | - Rui-Chuang Yang
- Research Center for Clinical and Translational Medicine, 302 Hospital of People’s Liberation ArmyBeijing, China
| | - Zhao-Fang Bai
- China Military Institute of Chinese Medicine, 302 Military Hospital of ChinaBeijing, China
| | - Li-Fu Wang
- Department of Integrative Medical Center, 302 Hospital of People’s Liberation ArmyBeijing, China
| | - Jian-Yu Li
- Department of Integrative Medical Center, 302 Hospital of People’s Liberation ArmyBeijing, China
| | - Hong-Hong Liu
- Department of Integrative Medical Center, 302 Hospital of People’s Liberation ArmyBeijing, China
| | - Yan-Ling Zhao
- Department of Pharmacy, 302 Military Hospital of ChinaBeijing, China
| | - Xiao-He Xiao
- China Military Institute of Chinese Medicine, 302 Military Hospital of ChinaBeijing, China
| |
Collapse
|
160
|
Sadallah S, Schmied L, Eken C, Charoudeh HN, Amicarella F, Schifferli JA. Platelet-Derived Ectosomes Reduce NK Cell Function. THE JOURNAL OF IMMUNOLOGY 2016; 197:1663-71. [DOI: 10.4049/jimmunol.1502658] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 06/21/2016] [Indexed: 11/19/2022]
|
161
|
Saunders PM, Vivian JP, O'Connor GM, Sullivan LC, Pymm P, Rossjohn J, Brooks AG. A bird's eye view of NK cell receptor interactions with their MHC class I ligands. Immunol Rev 2016; 267:148-66. [PMID: 26284476 DOI: 10.1111/imr.12319] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The surveillance of target cells by natural killer (NK) cells utilizes an ensemble of inhibitory and activating receptors, many of which interact with major histocompatibility complex (MHC) class I molecules. NK cell recognition of MHC class I proteins is important developmentally for the acquisition of full NK cell effector capacity and during target cell recognition, where the engagement of inhibitory receptors and MHC class I molecules attenuates NK cell activation. Human NK cells have evolved two broad strategies for recognition of human leukocyte antigen (HLA) class I molecules: (i) direct recognition of polymorphic classical HLA class I proteins by diverse receptor families such as the killer cell immunoglobulin-like receptors (KIRs), and (ii) indirect recognition of conserved sets of HLA class I-derived peptides displayed on the non-classical HLA-E for recognition by CD94-NKG2 receptors. In this review, we assess the structural basis for the interaction between these NK receptors and their HLA class I ligands and, using the suite of published KIR and CD94-NKG2 ternary complexes, highlight the features that allow NK cells to orchestrate the recognition of a range of different HLA class I proteins.
Collapse
Affiliation(s)
- Philippa M Saunders
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Julian P Vivian
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC, Australia
| | - Geraldine M O'Connor
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Lucy C Sullivan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Phillip Pymm
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC, Australia
| | - Jamie Rossjohn
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC, Australia.,Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Andrew G Brooks
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
162
|
Oszmiana A, Williamson DJ, Cordoba SP, Morgan DJ, Kennedy PR, Stacey K, Davis DM. The Size of Activating and Inhibitory Killer Ig-like Receptor Nanoclusters Is Controlled by the Transmembrane Sequence and Affects Signaling. Cell Rep 2016; 15:1957-72. [PMID: 27210755 PMCID: PMC4893158 DOI: 10.1016/j.celrep.2016.04.075] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 03/15/2016] [Accepted: 04/20/2016] [Indexed: 01/24/2023] Open
Abstract
Super-resolution microscopy has revealed that immune cell receptors are organized in nanoscale clusters at cell surfaces and immune synapses. However, mechanisms and functions for this nanoscale organization remain unclear. Here, we used super-resolution microscopy to compare the surface organization of paired killer Ig-like receptors (KIR), KIR2DL1 and KIR2DS1, on human primary natural killer cells and cell lines. Activating KIR2DS1 assembled in clusters two-fold larger than its inhibitory counterpart KIR2DL1. Site-directed mutagenesis established that the size of nanoclusters is controlled by transmembrane amino acid 233, a lysine in KIR2DS1. Super-resolution microscopy also revealed two ways in which the nanoscale clustering of KIR affects signaling. First, KIR2DS1 and DAP12 nanoclusters are juxtaposed in the resting cell state but coalesce upon receptor ligation. Second, quantitative super-resolution microscopy revealed that phosphorylation of the kinase ZAP-70 or phosphatase SHP-1 is favored in larger KIR nanoclusters. Thus, the size of KIR nanoclusters depends on the transmembrane sequence and affects downstream signaling. Activating and inhibitory NK cell receptors have a distinct nanoscale organization The transmembrane sequence of KIR controls their nanoscale organization Nanoclusters of KIR2DS1 and its adaptor are juxtaposed but mix upon activation Phosphorylation of ZAP-70 or SHP-1 is favored in larger receptor nanoclusters
Collapse
Affiliation(s)
- Anna Oszmiana
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - David J Williamson
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - Shaun-Paul Cordoba
- Division of Cell and Molecular Biology, Sir Alexander Fleming Building, Imperial College London, London SW7 2AZ, UK
| | - David J Morgan
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - Philippa R Kennedy
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - Kevin Stacey
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| | - Daniel M Davis
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK.
| |
Collapse
|
163
|
Varbanova V, Naumova E, Mihaylova A. Killer-cell immunoglobulin-like receptor genes and ligands and their role in hematologic malignancies. Cancer Immunol Immunother 2016; 65:427-40. [PMID: 26874942 PMCID: PMC11029164 DOI: 10.1007/s00262-016-1806-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 01/29/2016] [Indexed: 12/22/2022]
Abstract
Natural killer (NK) cells are considered crucial for the elimination of emerging tumor cells. Effector NK-cell functions are controlled by interactions of inhibitory and activating killer-cell immunoglobulin-like receptors (KIRs) on NK cells with human leukocyte antigen (HLA) class I ligands on target cells. KIR and HLA are highly polymorphic genetic systems segregating independently, creating a great diversity in KIR/HLA gene profiles in different individuals. There is an increasing evidence supporting the relevance of KIR and HLA ligand gene background for the occurrence and outcome of certain cancers. However, the data are still controversial and the mechanisms of receptor-ligand mediated NK-cell action remain unclear. Here, the main characteristics and functions of KIRs and their HLA class I ligands are reviewed. In addition, we review the HLA and KIR correlations with different hematological malignancies and discuss our current understanding of the biological significance and mechanisms underlying these associations.
Collapse
Affiliation(s)
- Viktoria Varbanova
- National Specialized Hospital for Active Treatment of Haematological Diseases, Sofia, Bulgaria
| | - Elissaveta Naumova
- Department of Clinical Immunology and Stem Cell Bank, University Hospital "Alexandrovska", Medical University, 1, Georgi Sofiiski Str., 1431, Sofia, Bulgaria
| | - Anastasiya Mihaylova
- Department of Clinical Immunology and Stem Cell Bank, University Hospital "Alexandrovska", Medical University, 1, Georgi Sofiiski Str., 1431, Sofia, Bulgaria.
| |
Collapse
|
164
|
Regulation of microglial survival and proliferation in health and diseases. Semin Immunol 2016; 27:410-5. [PMID: 27033414 DOI: 10.1016/j.smim.2016.03.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 03/16/2016] [Indexed: 11/23/2022]
Abstract
Microglia play an important role in the development and maintenance of the central nervous system (CNS) under homeostatic conditions as well as during neurodegenerative diseases. Recent observations in human genomics and advances in genetic mouse models have provided insights into signaling pathways that control development, survival, proliferation and function of microglia. Alteration of these pathways contributes to the pathogenesis of CNS diseases. Here we review the current literature regarding the roles of these microglial pathways in both the normal and diseased brain and discuss areas that require further investigation.
Collapse
|
165
|
Biochemical and Functional Insights into the Integrated Regulation of Innate Immune Cell Responses by Teleost Leukocyte Immune-Type Receptors. BIOLOGY 2016; 5:biology5010013. [PMID: 27005670 PMCID: PMC4810170 DOI: 10.3390/biology5010013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/01/2016] [Accepted: 03/01/2016] [Indexed: 12/22/2022]
Abstract
Across vertebrates, innate immunity consists of a complex assortment of highly specialized cells capable of unleashing potent effector responses designed to destroy or mitigate foreign pathogens. The execution of various innate cellular behaviors such as phagocytosis, degranulation, or cell-mediated cytotoxicity are functionally indistinguishable when being performed by immune cells isolated from humans or teleost fishes; vertebrates that diverged from one another more than 450 million years ago. This suggests that vital components of the vertebrate innate defense machinery are conserved and investigating such processes in a range of model systems provides an important opportunity to identify fundamental features of vertebrate immunity. One characteristic that is highly conserved across vertebrate systems is that cellular immune responses are dependent on specialized immunoregulatory receptors that sense environmental stimuli and initiate intracellular cascades that can elicit appropriate effector responses. A wide variety of immunoregulatory receptor families have been extensively studied in mammals, and many have been identified as cell- and function-specific regulators of a range of innate responses. Although much less is known in fish, the growing database of genomic information has recently allowed for the identification of several immunoregulatory receptor gene families in teleosts. Many of these putative immunoregulatory receptors have yet to be assigned any specific role(s), and much of what is known has been based solely on structural and/or phylogenetic relationships with mammalian receptor families. As an attempt to address some of these shortcomings, this review will focus on our growing understanding of the functional roles played by specific members of the channel catfish (Ictalurus punctatus) leukocyte immune-type receptors (IpLITRs), which appear to be important regulators of several innate cellular responses via classical as well as unique biochemical signaling networks.
Collapse
|
166
|
Abstract
NKG2D is an activating receptor expressed on the surface of natural killer (NK) cells, CD8(+) T cells, and subsets of CD4(+) T cells, invariant NKT cells (iNKT), and γδ T cells. In humans, NKG2D transmits signals by its association with the DAP10 adapter subunit, and in mice alternatively spliced isoforms transmit signals either using DAP10 or DAP12 adapter subunits. Although NKG2D is encoded by a highly conserved gene (KLRK1) with limited polymorphism, the receptor recognizes an extensive repertoire of ligands, encoded by at least eight genes in humans (MICA, MICB, RAET1E, RAET1G, RAET1H, RAET1I, RAET1L, and RAET1N), some with extensive allelic polymorphism. Expression of the NKG2D ligands is tightly regulated at the level of transcription, translation, and posttranslation. In general, healthy adult tissues do not express NKG2D glycoproteins on the cell surface, but these ligands can be induced by hyperproliferation and transformation, as well as when cells are infected by pathogens. Thus, the NKG2D pathway serves as a mechanism for the immune system to detect and eliminate cells that have undergone "stress." Viruses and tumor cells have devised numerous strategies to evade detection by the NKG2D surveillance system, and diversification of the NKG2D ligand genes likely has been driven by selective pressures imposed by pathogens. NKG2D provides an attractive target for therapeutics in the treatment of infectious diseases, cancer, and autoimmune diseases.
Collapse
Affiliation(s)
- Lewis L Lanier
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California.
| |
Collapse
|
167
|
Ancient Genetic Signatures of Orang Asli Revealed by Killer Immunoglobulin-Like Receptor Gene Polymorphisms. PLoS One 2015; 10:e0141536. [PMID: 26565719 PMCID: PMC4643969 DOI: 10.1371/journal.pone.0141536] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/10/2015] [Indexed: 02/06/2023] Open
Abstract
The aboriginal populations of Peninsular Malaysia, also known as Orang Asli (OA), comprise three major groups; Semang, Senoi and Proto-Malays. Here, we analyzed for the first time KIR gene polymorphisms for 167 OA individuals, including those from four smallest OA subgroups (Che Wong, Orang Kanaq, Lanoh and Kensiu) using polymerase chain reaction-sequence specific primer (PCR-SSP) analyses. The observed distribution of KIR profiles of OA is heterogenous; Haplotype B is the most frequent in the Semang subgroups (especially Batek) while Haplotype A is the most common type in the Senoi. The Semang subgroups were clustered together with the Africans, Indians, Papuans and Australian Aborigines in a principal component analysis (PCA) plot and shared many common genotypes (AB6, BB71, BB73 and BB159) observed in these other populations. Given that these populations also display high frequencies of Haplotype B, it is interesting to speculate that Haplotype B may be generally more frequent in ancient populations. In contrast, the two Senoi subgroups, Che Wong and Semai are displaced toward Southeast Asian and African populations in the PCA scatter plot, respectively. Orang Kanaq, the smallest and the most endangered of all OA subgroups, has lost some degree of genetic variation, as shown by their relatively high frequency of the AB2 genotype (0.73) and a total absence of KIR2DL2 and KIR2DS2 genes. Orang Kanaq tradition that strictly prohibits intermarriage with outsiders seems to have posed a serious threat to their survival. This present survey is a demonstration of the value of KIR polymorphisms in elucidating genetic relationships among human populations.
Collapse
|
168
|
Comparative study of various subpopulations of cytotoxic cells in blood and ascites from patients with ovarian carcinoma. Contemp Oncol (Pozn) 2015; 19:290-9. [PMID: 26557777 PMCID: PMC4631306 DOI: 10.5114/wo.2015.54388] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 11/10/2014] [Accepted: 03/18/2015] [Indexed: 12/13/2022] Open
Abstract
AIM OF THE STUDY A number of observations have indicated that the immune system plays a significant role in patients with epithelial ovarian cancer (EOC). In cases of EOC, the prognostic significance of tumour infiltrating lymphocytes has not been clearly explained yet. The aim is to determine the phenotype and activation molecules of cytotoxic T cell and NK cell subpopulations and to compare their representation in malignant ascites and peripheral blood in patients with ovarian cancer. MATERIAL AND METHODS Cytotoxic cells taken from blood samples of the cubital vein and malignant ascites were obtained from 53 patients with EOC. Their surface and activation characteristics were determined by means of a flow cytometer. Immunophenotype multiparametric analysis of peripheral blood lymphocytes (PBLs) and tumour infiltrating lymphocytes (TILs) was carried out. RESULTS CD3(+) T lymphocytes were the main population of TILs (75.9%) and PBLs (70.9%). The number of activating T cells was significantly higher in TILs: CD3(+)/69(+) 6.7% vs. 0.8% (p < 0.001). The representation of (CD3(-)/16(+)56(+)) NK cells in TILs was significantly higher: 11.0% vs. 5.6% (p = 0.041); likewise CD56(bright) and CD-56(bright) from CD56(+) cells were higher in TILs (both p < 0.001). The activation receptor NKG2D was present in 45.1% of TILs vs. 32.3% of PBLs (p = 0.034), but we did not find a significant difference in the numbers of CD56(+)/NKG2D(+) in TILs and PBLs. CONCLUSIONS These results prove that the characteristics and intensity of anti-tumour responses are different in compared compartments (ascites/PBLs). The knowledge of phenotype and functions of effector cells is the basic precondition for understanding the anti-tumour immune response.
Collapse
|
169
|
Painter MM, Atagi Y, Liu CC, Rademakers R, Xu H, Fryer JD, Bu G. TREM2 in CNS homeostasis and neurodegenerative disease. Mol Neurodegener 2015; 10:43. [PMID: 26337043 PMCID: PMC4560063 DOI: 10.1186/s13024-015-0040-9] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 08/19/2015] [Indexed: 02/07/2023] Open
Abstract
Myeloid-lineage cells accomplish a myriad of homeostatic tasks including the recognition of pathogens, regulation of the inflammatory milieu, and mediation of tissue repair and regeneration. The innate immune receptor and its adaptor protein—triggering receptor expressed on myeloid cells 2 (TREM2) and DNAX-activating protein of 12 kDa (DAP12)—possess the ability to modulate critical cellular functions via crosstalk with diverse signaling pathways. As such, mutations in TREM2 and DAP12 have been found to be associated with a range of disease phenotypes. In particular, mutations in TREM2 increase the risk for Alzheimer's disease and other neurodegenerative disorders. The leading hypothesis is that microglia, the resident immune cells of the central nervous system, are the major myeloid cells affected by dysregulated TREM2-DAP12 function. Here, we review how impaired signaling by the TREM2-DAP12 pathway leads to altered immune responses in phagocytosis, cytokine production, and microglial proliferation and survival, thus contributing to disease pathogenesis.
Collapse
Affiliation(s)
- Meghan M Painter
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| | - Yuka Atagi
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| | - Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA. .,Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| | - Huaxi Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
| | - John D Fryer
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA. .,Neurobiology of Disease Graduate Program, Mayo Clinic College of Medicine, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA. .,Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian, 361102, China. .,Neurobiology of Disease Graduate Program, Mayo Clinic College of Medicine, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| |
Collapse
|
170
|
TREM2 mRNA Expression in Leukocytes Is Increased in Alzheimer's Disease and Schizophrenia. PLoS One 2015; 10:e0136835. [PMID: 26332043 PMCID: PMC4557831 DOI: 10.1371/journal.pone.0136835] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 08/10/2015] [Indexed: 12/21/2022] Open
Abstract
TREM2 and TYROBP are causal genes for Nasu–Hakola disease (NHD), a rare autosomal recessive disease characterized by bone lesions and early-onset progressive dementia. TREM2 forms a receptor signaling complex with TYROBP, which triggers the activation of immune responses in macrophages and dendritic cells, and the functional polymorphism of TREM2 is reported to be associated with neurodegenerative disorders such as Alzheimer’s disease (AD). The objective of this study was to reveal the involvement of TYROBP and TREM2 in the pathophysiology of AD and schizophrenia. Methods: We investigated the mRNA expression level of the 2 genes in leukocytes of 26 patients with AD and 24 with schizophrenia in comparison with age-matched controls. Moreover, we performed gene association analysis between these 2 genes and schizophrenia. Results: No differences were found in TYROBP mRNA expression in patients with AD and schizophrenia; however, TREM2 mRNA expression was increased in patients with AD and schizophrenia compared with controls (P < 0.001). There were no genetic associations of either gene with schizophrenia in Japanese patients. Conclusion: TREM2 expression in leukocytes is elevated not only in AD but also in schizophrenia. Inflammatory processes involving TREM2 may occur in schizophrenia, as observed in neurocognitive disorders such as AD. TREM2 expression in leukocytes may be a novel biomarker for neurological and psychiatric disorders.
Collapse
|
171
|
Radjabova V, Mastroeni P, Skjødt K, Zaccone P, de Bono B, Goodall JC, Chilvers ER, Juss JK, Jones DC, Trowsdale J, Barrow AD. TARM1 Is a Novel Leukocyte Receptor Complex-Encoded ITAM Receptor That Costimulates Proinflammatory Cytokine Secretion by Macrophages and Neutrophils. THE JOURNAL OF IMMUNOLOGY 2015; 195:3149-59. [PMID: 26311901 DOI: 10.4049/jimmunol.1401847] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 07/19/2015] [Indexed: 01/22/2023]
Abstract
We identified a novel, evolutionarily conserved receptor encoded within the human leukocyte receptor complex and syntenic region of mouse chromosome 7, named T cell-interacting, activating receptor on myeloid cells-1 (TARM1). The transmembrane region of TARM1 contained a conserved arginine residue, consistent with association with a signaling adaptor. TARM1 associated with the ITAM adaptor FcRγ but not with DAP10 or DAP12. In healthy mice, TARM1 is constitutively expressed on the cell surface of mature and immature CD11b(+)Gr-1(+) neutrophils within the bone marrow. Following i.p. LPS treatment or systemic bacterial challenge, TARM1 expression was upregulated by neutrophils and inflammatory monocytes and TARM1(+) cells were rapidly recruited to sites of inflammation. TARM1 expression was also upregulated by bone marrow-derived macrophages and dendritic cells following stimulation with TLR agonists in vitro. Ligation of TARM1 receptor in the presence of TLR ligands, such as LPS, enhanced the secretion of proinflammatory cytokines by macrophages and primary mouse neutrophils, whereas TARM1 stimulation alone had no effect. Finally, an immobilized TARM1-Fc fusion protein suppressed CD4(+) T cell activation and proliferation in vitro. These results suggest that a putative T cell ligand can interact with TARM1 receptor, resulting in bidirectional signaling and raising the T cell activation threshold while costimulating the release of proinflammatory cytokines by macrophages and neutrophils.
Collapse
Affiliation(s)
- Valeria Radjabova
- Division of Immunology, Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom
| | - Piero Mastroeni
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| | - Karsten Skjødt
- Department of Cancer and Inflammation, Institute for Molecular Medicine, University of Southern Denmark, DK-5000 Odense, Denmark
| | - Paola Zaccone
- Division of Immunology, Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom
| | - Bernard de Bono
- Centre for Health Informatics and Multiprofessional Education, University College London, London NW1 2DA, United Kingdom
| | - Jane C Goodall
- Department of Medicine, University of Cambridge, Cambridge CB2 0SP, United Kingdom; and
| | - Edwin R Chilvers
- Department of Medicine, University of Cambridge, Cambridge CB2 0SP, United Kingdom; and
| | - Jatinder K Juss
- Department of Medicine, University of Cambridge, Cambridge CB2 0SP, United Kingdom; and
| | - Des C Jones
- Division of Immunology, Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom
| | - John Trowsdale
- Division of Immunology, Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom;
| | - Alexander David Barrow
- Division of Immunology, Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
172
|
Hilton HG, Guethlein LA, Goyos A, Nemat-Gorgani N, Bushnell DA, Norman PJ, Parham P. Polymorphic HLA-C Receptors Balance the Functional Characteristics of KIR Haplotypes. THE JOURNAL OF IMMUNOLOGY 2015; 195:3160-70. [PMID: 26311903 DOI: 10.4049/jimmunol.1501358] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 07/22/2015] [Indexed: 01/04/2023]
Abstract
The human killer cell Ig-like receptor (KIR) locus comprises two groups of KIR haplotypes, termed A and B. These are present in all human populations but with different relative frequencies, suggesting they have different functional properties that underlie their balancing selection. We studied the genomic organization and functional properties of the alleles of the inhibitory and activating HLA-C receptors encoded by KIR haplotypes. Because every HLA-C allotype functions as a ligand for KIR, the interactions between KIR and HLA-C dominate the HLA class I-mediated regulation of human NK cells. The C2 epitope is recognized by inhibitory KIR2DL1 and activating KIR2DS1, whereas the C1 epitope is recognized by inhibitory KIR2DL2 and KIR2DL3. This study shows that the KIR2DL1, KIR2DS1, and KIR2DL2/3 alleles form distinctive phylogenetic clades that associate with specific KIR haplotypes. KIR A haplotypes are characterized by KIR2DL1 alleles that encode strong inhibitory C2 receptors and KIR2DL3 alleles encoding weak inhibitory C1 receptors. In striking contrast, KIR B haplotypes are characterized by KIR2DL1 alleles that encode weak inhibitory C2 receptors and KIR2DL2 alleles encoding strong inhibitory C1 receptors. The wide-ranging properties of KIR allotypes arise from substitutions throughout the KIR molecule. Such substitutions can influence cell surface expression, as well as the avidity and specificity for HLA-C ligands. Consistent with the crucial role of inhibitory HLA-C receptors in self-recognition, as well as NK cell education and response, most KIR haplotypes have both a functional C1 and C2 receptor, despite the considerable variation that occurs in ligand recognition and surface expression.
Collapse
Affiliation(s)
- Hugo G Hilton
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305; and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Lisbeth A Guethlein
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305; and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Ana Goyos
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305; and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Neda Nemat-Gorgani
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305; and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - David A Bushnell
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305; and
| | - Paul J Norman
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305; and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Peter Parham
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305; and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
173
|
Wang E, Wang LC, Tsai CY, Bhoj V, Gershenson Z, Moon E, Newick K, Sun J, Lo A, Baradet T, Feldman MD, Barrett D, Puré E, Albelda S, Milone MC. Generation of Potent T-cell Immunotherapy for Cancer Using DAP12-Based, Multichain, Chimeric Immunoreceptors. Cancer Immunol Res 2015; 3:815-26. [PMID: 25941351 PMCID: PMC4490943 DOI: 10.1158/2326-6066.cir-15-0054] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/14/2015] [Indexed: 11/16/2022]
Abstract
Chimeric antigen receptors (CAR) bearing an antigen-binding domain linked in cis to the cytoplasmic domains of CD3ζ and costimulatory receptors have provided a potent method for engineering T-cell cytotoxicity toward B-cell leukemia and lymphoma. However, resistance to immunotherapy due to loss of T-cell effector function remains a significant barrier, especially in solid malignancies. We describe an alternative chimeric immunoreceptor design in which we have fused a single-chain variable fragment for antigen recognition to the transmembrane and cytoplasmic domains of KIR2DS2, a stimulatory killer immunoglobulin-like receptor (KIR). We show that this simple, KIR-based CAR (KIR-CAR) triggers robust antigen-specific proliferation and effector function in vitro when introduced into human T cells with DAP12, an immunotyrosine-based activation motifs-containing adaptor. T cells modified to express a KIR-CAR and DAP12 exhibit superior antitumor activity compared with standard first- and second-generation CD3ζ-based CARs in a xenograft model of mesothelioma highly resistant to immunotherapy. The enhanced antitumor activity is associated with improved retention of chimeric immunoreceptor expression and improved effector function of isolated tumor-infiltrating lymphocytes. These results support the exploration of KIR-CARs for adoptive T-cell immunotherapy, particularly in immunotherapy-resistant solid tumors.
Collapse
Affiliation(s)
- Enxiu Wang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Liang-Chuan Wang
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ching-Yi Tsai
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Vijay Bhoj
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Zack Gershenson
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Edmund Moon
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kheng Newick
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jing Sun
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Albert Lo
- Department of Animal Biology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania
| | - Timothy Baradet
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael D Feldman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - David Barrett
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Ellen Puré
- Department of Animal Biology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania
| | - Steven Albelda
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael C Milone
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
174
|
Deng H, Zheng W, Jankovic J. Genetics and molecular biology of brain calcification. Ageing Res Rev 2015; 22:20-38. [PMID: 25906927 DOI: 10.1016/j.arr.2015.04.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 04/14/2015] [Accepted: 04/15/2015] [Indexed: 01/01/2023]
Abstract
Brain calcification is a common neuroimaging finding in patients with neurological, metabolic, or developmental disorders, mitochondrial diseases, infectious diseases, traumatic or toxic history, as well as in otherwise normal older people. Patients with brain calcification may exhibit movement disorders, seizures, cognitive impairment, and a variety of other neurologic and psychiatric symptoms. Brain calcification may also present as a single, isolated neuroimaging finding. When no specific cause is evident, a genetic etiology should be considered. The aim of the review is to highlight clinical disorders associated with brain calcification and provide summary of current knowledge of diagnosis, genetics, and pathogenesis of brain calcification.
Collapse
Affiliation(s)
- Hao Deng
- Department of Neurology, Third Xiangya Hospital, Central South University, Changsha, China; Center for Experimental Medicine, Third Xiangya Hospital, Central South University, Changsha, China.
| | - Wen Zheng
- Department of Neurology, Third Xiangya Hospital, Central South University, Changsha, China; Center for Experimental Medicine, Third Xiangya Hospital, Central South University, Changsha, China
| | - Joseph Jankovic
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
175
|
Sasaki A, Kakita A, Yoshida K, Konno T, Ikeuchi T, Hayashi S, Matsuo H, Shioda K. Variable expression of microglial DAP12 and TREM2 genes in Nasu-Hakola disease. Neurogenetics 2015; 16:265-76. [DOI: 10.1007/s10048-015-0451-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 05/10/2015] [Indexed: 11/29/2022]
|
176
|
Transmembrane Complexes of DAP12 Crystallized in Lipid Membranes Provide Insights into Control of Oligomerization in Immunoreceptor Assembly. Cell Rep 2015; 11:1184-92. [PMID: 25981043 DOI: 10.1016/j.celrep.2015.04.045] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/09/2015] [Accepted: 04/21/2015] [Indexed: 11/20/2022] Open
Abstract
The membrane-spanning α helices of single-pass receptors play crucial roles in stabilizing oligomeric structures and transducing biochemical signals across the membrane. Probing intermolecular transmembrane interactions in single-pass receptors presents unique challenges, reflected in a gross underrepresentation of their membrane-embedded domains in structural databases. Here, we present two high-resolution structures of transmembrane assemblies from a eukaryotic single-pass protein crystallized in a lipidic membrane environment. Trimeric and tetrameric structures of the immunoreceptor signaling module DAP12, determined to 1.77-Å and 2.14-Å resolution, respectively, are organized by the same polar surfaces that govern intramembrane assembly with client receptors. We demonstrate that, in addition to the well-studied dimeric form, these trimeric and tetrameric structures are made in cells, and their formation is competitive with receptor association in the ER. The polar transmembrane sequences therefore act as primary determinants of oligomerization specificity through interplay between charge shielding and sequestration of polar surfaces within helix interfaces.
Collapse
|
177
|
Zhong L, Chen XF, Zhang ZL, Wang Z, Shi XZ, Xu K, Zhang YW, Xu H, Bu G. DAP12 Stabilizes the C-terminal Fragment of the Triggering Receptor Expressed on Myeloid Cells-2 (TREM2) and Protects against LPS-induced Pro-inflammatory Response. J Biol Chem 2015; 290:15866-15877. [PMID: 25957402 DOI: 10.1074/jbc.m115.645986] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Indexed: 11/06/2022] Open
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM2) is a DAP12-associated receptor expressed in microglia, macrophages, and other myeloid-derived cells. Previous studies have suggested that TREM2/DAP12 signaling pathway reduces inflammatory responses and promotes phagocytosis of apoptotic neurons. Recently, TREM2 has been identified as a risk gene for Alzheimer disease (AD). Here, we show that DAP12 stabilizes the C-terminal fragment of TREM2 (TREM2-CTF), a substrate for γ-secretase. Co-expression of DAP12 with TREM2 selectively increased the level of TREM2-CTF with little effects on that of full-length TREM2. The interaction between DAP12 and TREM2 is essential for TREM2-CTF stabilization as a mutant form of DAP12 with disrupted interaction with TREM2 failed to exhibit such an effect. Silencing of either Trem2 or Dap12 gene significantly exacerbated pro-inflammatory responses induced by lipopolysaccharides (LPS). Importantly, overexpression of either full-length TREM2 or TREM2-CTF reduced LPS-induced inflammatory responses. Taken together, our results support a role of DAP12 in stabilizing TREM2-CTF, thereby protecting against excessive pro-inflammatory responses.
Collapse
Affiliation(s)
- Li Zhong
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen 361102, PR China
| | - Xiao-Fen Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen 361102, PR China.
| | - Zhen-Lian Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen 361102, PR China
| | - Zhe Wang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen 361102, PR China
| | - Xin-Zhen Shi
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen 361102, PR China
| | - Kai Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen 361102, PR China
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen 361102, PR China
| | - Huaxi Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen 361102, PR China; Degenerative Disease Research Program, Center for Neuroscience, Aging, and Stem Cell Research, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Guojun Bu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen 361102, PR China; Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224.
| |
Collapse
|
178
|
Hernangómez M, Carrillo-Salinas FJ, Mecha M, Correa F, Mestre L, Loría F, Feliú A, Docagne F, Guaza C. Brain innate immunity in the regulation of neuroinflammation: therapeutic strategies by modulating CD200-CD200R interaction involve the cannabinoid system. Curr Pharm Des 2015; 20:4707-22. [PMID: 24588829 PMCID: PMC4157566 DOI: 10.2174/1381612820666140130202911] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 01/29/2014] [Indexed: 11/24/2022]
Abstract
The central nervous system (CNS) innate immune response includes an arsenal of molecules and receptors expressed by professional phagocytes, glial cells and neurons that is involved in host defence and clearance of toxic and dangerous cell debris. However, any uncontrolled innate immune responses within the CNS are widely recognized as playing a major role in the development of autoimmune disorders and neurodegeneration, with multiple sclerosis (MS) Alzheimer's disease (AD) being primary examples. Hence, it is important to identify the key regulatory mechanisms involved in the control of CNS innate immunity and which could be harnessed to explore novel therapeutic avenues. Neuroimmune regulatory proteins (NIReg) such as CD95L, CD200, CD47, sialic acid, complement regulatory proteins (CD55, CD46, fH, C3a), HMGB1, may control the adverse immune responses in health and diseases. In the absence of these regulators, when neurons die by apoptosis, become infected or damaged, microglia and infiltrating immune cells are free to cause injury as well as an adverse inflammatory response in acute and chronic settings. We will herein provide new emphasis on the role of the pair CD200-CD200R in MS and its experimental models: experimental autoimmune encephalomyelitis (EAE) and Theiler’s virus induced demyelinating disease (TMEV-IDD). The interest of the cannabinoid system as inhibitor of inflammation prompt us to introduce our findings about the role of endocannabinoids (eCBs) in promoting CD200-CD200 receptor (CD200R) interaction and the benefits caused in TMEV-IDD. Finally, we also review the current data on CD200-CD200R interaction in AD, as well as, in the aging brain.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Carmen Guaza
- Neuroimmunology Group, Functional and Systems Neurobiology Department, Instituto Cajal, CSIC, 28002 Madrid, Spain.
| |
Collapse
|
179
|
Xing J, Titus AR, Humphrey MB. The TREM2-DAP12 signaling pathway in Nasu-Hakola disease: a molecular genetics perspective. ACTA ACUST UNITED AC 2015; 5:89-100. [PMID: 26478868 PMCID: PMC4605443 DOI: 10.2147/rrbc.s58057] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nasu–Hakola disease or polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy (PLOSL) is a rare recessively inherited disease that is associated with early dementia and bone cysts with fractures. Here, we review the genetic causes of PLOSL with loss-of-function mutations or deletions in one of two genes, TYROBP and TREM2, encoding for two proteins DNAX-activating protein 12 (DAP12) and triggering receptor expressed on myeloid cells-2 (TREM2). TREM2 and DAP12 form an immunoreceptor signaling complex that mediates myeloid cell, including microglia and osteoclasts, development, activation, and function. Functionally, TREM2-DAP12 mediates osteoclast multi-nucleation, migration, and resorption. In microglia, TREM2-DAP12 participates in recognition and apoptosis of neuronal debris and amyloid deposits. Review of the complex immunoregulatory roles of TREM2-DAP12 in the innate immune system, where it can both promote and inhibit pro-inflammatory responses, is given. Little is known about the function of TREM2-DAP12 in normal brain homeostasis or in pathological central nervous system diseases. Based on the state of the field, genetic testing now aids in diagnosis of PLOSL, but therapeutics and interventions are still under development.
Collapse
Affiliation(s)
- Junjie Xing
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA ; Department of Microbiology and immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Amanda R Titus
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Mary Beth Humphrey
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA ; Department of Microbiology and immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA ; Department of veteran's Affairs, Oklahoma City, OK, USA
| |
Collapse
|
180
|
Töpfer K, Cartellieri M, Michen S, Wiedemuth R, Müller N, Lindemann D, Bachmann M, Füssel M, Schackert G, Temme A. DAP12-based activating chimeric antigen receptor for NK cell tumor immunotherapy. THE JOURNAL OF IMMUNOLOGY 2015; 194:3201-12. [PMID: 25740942 DOI: 10.4049/jimmunol.1400330] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
NK cells are emerging as new effectors for immunotherapy of cancer. In particular, the genetic engraftment of chimeric Ag receptors (CARs) in NK cells is a promising strategy to redirect NK cells to otherwise NK cell-resistant tumor cells. On the basis of DNAX-activation protein 12 (DAP12), a signaling adaptor molecule involved in signal transduction of activating NK cell receptors, we generated a new type of CAR targeting the prostate stem cell Ag (PSCA). We demonstrate in this article that this CAR, designated anti-PSCA-DAP12, consisting of DAP12 fused to the anti-PSCA single-chain Ab fragment scFv(AM1) confers improved cytotoxicity to the NK cell line YTS against PSCA-positive tumor cells when compared with a CAR containing the CD3ζ signaling chain. Further analyses revealed phosphorylation of the DAP12-associated ZAP-70 kinase and IFN-γ release of CAR-engineered cells after contact with PSCA-positive target cells. YTS cells modified with DAP12 alone or with a CAR bearing a phosphorylation-defective ITAM were not activated. Notably, infused YTS cells armed with anti-PSCA-DAP12 caused delayed tumor xenograft growth and resulted in complete tumor eradication in a significant fraction of treated mice. The feasibility of the DAP12-based CAR was further tested in human primary NK cells and confers specific cytotoxicity against KIR/HLA-matched PSCA-positive tumor cells, which was further enhanced by KIR-HLA mismatches. We conclude that NK cells engineered with DAP12-based CARs are a promising tool for adoptive tumor immunotherapy.
Collapse
Affiliation(s)
- Katrin Töpfer
- Section of Experimental Neurosurgery and Tumor Immunology, Department of Neurosurgery, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Marc Cartellieri
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany
| | - Susanne Michen
- Section of Experimental Neurosurgery and Tumor Immunology, Department of Neurosurgery, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Ralf Wiedemuth
- Section of Experimental Neurosurgery and Tumor Immunology, Department of Neurosurgery, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Nadja Müller
- Section of Experimental Neurosurgery and Tumor Immunology, Department of Neurosurgery, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Dirk Lindemann
- Institute of Virology, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany
| | - Monika Füssel
- DKMS Life Science Lab, GmbH, 01307 Dresden, Germany; and
| | - Gabriele Schackert
- Section of Experimental Neurosurgery and Tumor Immunology, Department of Neurosurgery, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Achim Temme
- Section of Experimental Neurosurgery and Tumor Immunology, Department of Neurosurgery, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; German Cancer Consortium (DKTK), 01307 Dresden, Germany
| |
Collapse
|
181
|
Zhang J, Basher F, Wu JD. NKG2D Ligands in Tumor Immunity: Two Sides of a Coin. Front Immunol 2015; 6:97. [PMID: 25788898 PMCID: PMC4349182 DOI: 10.3389/fimmu.2015.00097] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/18/2015] [Indexed: 11/13/2022] Open
Abstract
The activating/co-stimulatory receptor NKG2D (natural-killer group 2, member D) is expressed on the surface of all human NK, NKT, CD8(+) T, and subsets of γδ(+) T cells. The significance of NKG2D function in tumor immunity has been well demonstrated in experimental animal models. However, the role of human NKG2D ligands in regulating tumor immunity and cancer prognosis had been controversial in the literature. In this review, we summarize the latest advancement, discuss the controversies, and present evidence that membrane-bound and soluble NKG2D ligands oppositely regulate tumor immunity. We also discuss new perspectives of targeting NKG2D ligands for cancer immunotherapy.
Collapse
Affiliation(s)
- Jinyu Zhang
- Department of Microbiology and Immunology, Medical University of South Carolina , Charleston, SC , USA
| | - Fahmin Basher
- Department of Microbiology and Immunology, Medical University of South Carolina , Charleston, SC , USA
| | - Jennifer D Wu
- Department of Microbiology and Immunology, Medical University of South Carolina , Charleston, SC , USA ; Cancer Immunology Program, Hollings Cancer Center , Charleston, SC , USA
| |
Collapse
|
182
|
Mbongue JC, Nicholas DA, Zhang K, Kim NS, Hamilton BN, Larios M, Zhang G, Umezawa K, Firek AF, Langridge WHR. Induction of indoleamine 2, 3-dioxygenase in human dendritic cells by a cholera toxin B subunit-proinsulin vaccine. PLoS One 2015; 10:e0118562. [PMID: 25714914 PMCID: PMC4340906 DOI: 10.1371/journal.pone.0118562] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 01/20/2015] [Indexed: 12/28/2022] Open
Abstract
Dendritic cells (DC) interact with naïve T cells to regulate the delicate balance between immunity and tolerance required to maintain immunological homeostasis. In this study, immature human dendritic cells (iDC) were inoculated with a chimeric fusion protein vaccine containing the pancreatic β-cell auto-antigen proinsulin linked to a mucosal adjuvant the cholera toxin B subunit (CTB-INS). Proteomic analysis of vaccine inoculated DCs revealed strong up-regulation of the tryptophan catabolic enzyme indoleamine 2, 3-dioxygenase (IDO1). Increased biosynthesis of the immunosuppressive enzyme was detected in DCs inoculated with the CTB-INS fusion protein but not in DCs inoculated with proinsulin, CTB, or an unlinked combination of the two proteins. Immunoblot and PCR analyses of vaccine treated DCs detected IDO1mRNA by 3 hours and IDO1 protein synthesis by 6 hours after vaccine inoculation. Determination of IDO1 activity in vaccinated DCs by measurement of tryptophan degradation products (kynurenines) showed increased tryptophan cleavage into N-formyl kynurenine. Vaccination did not interfere with monocytes differentiation into DC, suggesting the vaccine can function safely in the human immune system. Treatment of vaccinated DCs with pharmacological NF-κB inhibitors ACHP or DHMEQ significantly inhibited IDO1 biosynthesis, suggesting a role for NF-κB signaling in vaccine up-regulation of dendritic cell IDO1. Heat map analysis of the proteomic data revealed an overall down-regulation of vaccinated DC functions, suggesting vaccine suppression of DC maturation. Together, our experimental data indicate that CTB-INS vaccine induction of IDO1 biosynthesis in human DCs may result in the inhibition of DC maturation generating a durable state of immunological tolerance. Understanding how CTB-INS modulates IDO1 activity in human DCs will facilitate vaccine efficacy and safety, moving this immunosuppressive strategy closer to clinical applications for prevention of type 1 diabetes autoimmunity.
Collapse
Affiliation(s)
- Jacques C. Mbongue
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
- Loma Linda University School of Medicine, Department of Basic Sciences, Division of Physiology, Loma Linda, CA, United States of America
| | - Dequina A. Nicholas
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
- Mass Spectrometer Core Facility, Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda University School of Medicine, Department of Basic Sciences, Loma Linda, CA, United States of America
| | - Kangling Zhang
- Mass Spectrometer Core Facility, Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda University School of Medicine, Department of Basic Sciences, Loma Linda, CA, United States of America
- Department of Pharmacology and Toxicology, School of Medicine, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Nan-Sun Kim
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
- Mass Spectrometer Core Facility, Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda University School of Medicine, Department of Basic Sciences, Loma Linda, CA, United States of America
- Department of Molecular Biology, Chonbuk National University, Jeon-Ju, Republic of Korea
| | - Brittany N. Hamilton
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
- Loma Linda University School of Medicine, Department of Basic Sciences, Division of Microbiology and Molecular Genetics, Loma Linda, CA, United States of America
| | - Marco Larios
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
| | - Guangyu Zhang
- Mass Spectrometer Core Facility, Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda University School of Medicine, Department of Basic Sciences, Loma Linda, CA, United States of America
| | - Kazuo Umezawa
- Aichi Medical University, School of Medicine, Department of Molecular Target Medicine Screening, Nagakute, Aichi, Japan
| | - Anthony F. Firek
- Endocrinology Section, JL Pettis Memorial VA Medical Center, Loma Linda, CA, United States of America
| | - William H. R. Langridge
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States of America
- Mass Spectrometer Core Facility, Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda University School of Medicine, Department of Basic Sciences, Loma Linda, CA, United States of America
- * E-mail:
| |
Collapse
|
183
|
Foley B, Felices M, Cichocki F, Cooley S, Verneris MR, Miller JS. The biology of NK cells and their receptors affects clinical outcomes after hematopoietic cell transplantation (HCT). Immunol Rev 2015; 258:45-63. [PMID: 24517425 DOI: 10.1111/imr.12157] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Natural killer (NK) cells were first identified for their capacity to reject bone marrow allografts in lethally irradiated mice without prior sensitization. Subsequently, human NK cells were detected and defined by their non-major histocompatibility complex (MHC)-restricted cytotoxicity toward transformed or virally infected target cells. Karre et al. later proposed 'the missing self hypothesis' to explain the mechanism by which self-tolerant cells could kill targets that had lost self MHC class I. Subsequently, the receptors that recognize MHC class I to mediate tolerance in the host were identified on NK cells. These class I-recognizing receptors contribute to the acquisition of function by a dynamic process known as NK cell education or licensing. In the past, NK cells were assumed to be short lived, but more recently NK cells have been shown to mediate immunologic memory to secondary exposures to cytomegalovirus infection. Because of their ability to lyse tumors with aberrant MHC class I expression and to produce cytokines and chemokines upon activation, NK cells may be primed by many stimuli, including viruses and inflammation, to contribute to a graft-versus-tumor effect. In addition, interactions with other immune cells support the therapeutic potential of NK cells to eradicate tumor and to enhance outcomes after hematopoietic cell transplantation.
Collapse
Affiliation(s)
- Bree Foley
- Adult, Oncology and Transplantation, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN, USA
| | - Martin Felices
- Adult, Oncology and Transplantation, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN, USA
| | - Frank Cichocki
- Adult, Oncology and Transplantation, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN, USA
| | - Sarah Cooley
- Adult, Oncology and Transplantation, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN, USA
| | - Michael R Verneris
- Pediatric Hematology, Oncology and Transplantation, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN, USA
| | - Jeffrey S Miller
- Adult, Oncology and Transplantation, Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
184
|
Liu Y, Liu J, Pang X, Liu T, Ning Z, Cheng G. The roles of direct recognition by animal lectins in antiviral immunity and viral pathogenesis. Molecules 2015; 20:2272-95. [PMID: 25642837 PMCID: PMC6272511 DOI: 10.3390/molecules20022272] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 01/21/2015] [Indexed: 11/24/2022] Open
Abstract
Lectins are a group of proteins with carbohydrate recognition activity. Lectins are categorized into many families based on their different cellular locations as well as their specificities for a variety of carbohydrate structures due to the features of their carbohydrate recognition domain (CRD) modules. Many studies have indicated that the direct recognition of particular oligosaccharides on viral components by lectins is important for interactions between hosts and viruses. Herein, we aim to globally review the roles of this recognition by animal lectins in antiviral immune responses and viral pathogenesis. The different classes of mammalian lectins can either recognize carbohydrates to activate host immunity for viral elimination or can exploit those carbohydrates as susceptibility factors to facilitate viral entry, replication or assembly. Additionally, some arthropod C-type lectins were recently identified as key susceptibility factors that directly interact with multiple viruses and then facilitate infection. Summarization of the pleiotropic roles of direct viral recognition by animal lectins will benefit our understanding of host-virus interactions and could provide insight into the role of lectins in antiviral drug and vaccine development.
Collapse
Affiliation(s)
- Yang Liu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China.
| | - Jianying Liu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China.
| | - Xiaojing Pang
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China.
| | - Tao Liu
- Center for Reproductive Medicine, Tai'an Central Hospital, Tai'an 271000, China.
| | - Zhijie Ning
- Ji'nan Infectious Diseases Hospital, Ji'nan 250021, China.
| | - Gong Cheng
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
185
|
Abstract
OBJECTIVES Streptococcus pneumoniae is the most common causative organism in community-acquired pneumonia responsible for millions of deaths every year. DNAX-activating protein of 12 kDa is an adaptor molecule for different myeloid expressed receptors involved in innate immunity. DESIGN Animal study. SETTING University research laboratory. SUBJECTS DNAX-activating protein of 12 kDa-deficient (dap12) and wild-type mice. INTERVENTIONS Mice were intranasally infected with S. pneumoniae. In addition, ex vivo responsiveness of alveolar macrophages was examined. MEASUREMENTS AND MAIN RESULTS dap12 alveolar macrophages released more tumor necrosis factor-α upon stimulation with S. pneumoniae and displayed increased phagocytosis of this pathogen compared with wild-type cells. After infection with S. pneumoniae via the airways, dap12 mice demonstrated reduced bacterial outgrowth in the lungs together with delayed dissemination to distant body sites relative to wild-type mice. This favorable response in dap12 mice was accompanied by reduced lung inflammation and an improved survival. CONCLUSIONS These data suggest that DNAX-activating protein of 12 kDa impairs host defense during pneumococcal pneumonia at the primary site of infection at least in part by inhibiting phagocytosis by alveolar macrophages.
Collapse
|
186
|
Cichocki F, Verneris MR, Cooley S, Bachanova V, Brunstein CG, Blazar BR, Wagner J, Schlums H, Bryceson YT, Weisdorf DJ, Miller JS. The Past, Present, and Future of NK Cells in Hematopoietic Cell Transplantation and Adoptive Transfer. Curr Top Microbiol Immunol 2015; 395:225-43. [PMID: 26037048 DOI: 10.1007/82_2015_445] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hematopoietic cell transplantation (HCT) has been used as a part of cancer therapy for over half a decade. Beyond the necessity for donor-derived cells to reconstitute hematopoiesis after radiation and chemotherapy, immunologic reconstitution from allogeneic cells is important for the elimination of residual tumor cells. Natural killer (NK) cells are first among lymphocytes to reconstitute post-transplant and protect against cancer relapse. In this review, we provide a historical perspective on the role of NK cells in cancer control in the transplant setting and focus on current research aimed at improving NK cell responses for therapeutic benefit.
Collapse
Affiliation(s)
- Frank Cichocki
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | | | - Sarah Cooley
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | | | | | - Bruce R Blazar
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - John Wagner
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Heinrich Schlums
- Centre for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Yenan T Bryceson
- Centre for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden.,Broeglmann Research Laboratory, Clinical Institute, University of Bergen, Bergen, Norway
| | - Daniel J Weisdorf
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Jeffrey S Miller
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA. .,MMC 806, Division of Hematology, Oncology and Transplantation, University of Minnesota Cancer Center, Harvard Street at East River Road, Minneapolis, MN, 55455, USA.
| |
Collapse
|
187
|
Lou Q, Zhang W, Liu G, Ma Y. The C-type lectin OCILRP2 costimulates EL4 T cell activation via the DAP12-Raf-MAP kinase pathway. PLoS One 2014; 9:e113218. [PMID: 25411776 PMCID: PMC4239057 DOI: 10.1371/journal.pone.0113218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 10/21/2014] [Indexed: 11/30/2022] Open
Abstract
OCILRP2 is a typical Type-II transmembrane protein that is selectively expressed in activated T lymphocytes, dendritic cells, and B cells and functions as a novel co-stimulator of T cell activation. However, the signaling pathways underlying OCILRP2 in T cell activation are still not completely understood. In this study, we found that the knockdown of OCILRP2 expression with shRNA or the blockage of its activity by an anti-OCILRP2 antagonist antibody reduced CD3/CD28-costimulated EL4 T cell viability and IL-2 production, inhibit Raf1, MAPK3, and MAPK8 activation, and impair NFAT and NF-κB transcriptional activities. Furthermore, immunoprecipitation results indicated that OCILRP2 could interact with the DAP12 protein, an adaptor containing an intracellular ITAM motif that can transduce signals to induce MAP kinase activation for T cell activation. Our data reveal that after binding with DAP12, OCILRP2 activates the Raf-MAP kinase pathways, resulting in T cell activation.
Collapse
Affiliation(s)
- Qiang Lou
- Henan Engineering Lab of Antibody Medicine, Key Laboratory of Cellular and Molecular Immunology, Medical College of Henan University, Kaifeng 475004, China
| | - Wei Zhang
- Henan Engineering Lab of Antibody Medicine, Key Laboratory of Cellular and Molecular Immunology, Medical College of Henan University, Kaifeng 475004, China
| | - Guangchao Liu
- Henan Engineering Lab of Antibody Medicine, Key Laboratory of Cellular and Molecular Immunology, Medical College of Henan University, Kaifeng 475004, China
| | - Yuanfang Ma
- Henan Engineering Lab of Antibody Medicine, Key Laboratory of Cellular and Molecular Immunology, Medical College of Henan University, Kaifeng 475004, China
- * E-mail:
| |
Collapse
|
188
|
Takeda K, Qin SY, Matsumoto N, Yamamoto K. Association of malectin with ribophorin I is crucial for attenuation of misfolded glycoprotein secretion. Biochem Biophys Res Commun 2014; 454:436-40. [DOI: 10.1016/j.bbrc.2014.10.102] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 10/20/2014] [Indexed: 10/24/2022]
|
189
|
Pelham CJ, Pandya AN, Agrawal DK. Triggering receptor expressed on myeloid cells receptor family modulators: a patent review. Expert Opin Ther Pat 2014; 24:1383-95. [PMID: 25363248 DOI: 10.1517/13543776.2014.977865] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Triggering receptor expressed on myeloid cells (TREM) receptors and TREM-like transcript (TLT; or TREML) receptors of the immunoglobulin superfamily are known as key modulators of host immune responses. TREM-1 (CD354) and TREM-2 share the transmembrane adaptor DNAX-activation protein of 12 kDa (DAP12), but they possess separate stimulatory and inhibitory functional roles, especially in myeloid cells. AREAS COVERED This review covers findings related to TREMs and TLTs published in patent applications from their discovery in 2000 to the present. New roles for TREM-1, TREM-2, TLT-1 and TLT-2 in maladies ranging from acute and chronic inflammatory disorders to cardiovascular diseases and cancers are discussed. Putative endogenous ligands and novel synthetic peptide blockers are also discussed. EXPERT OPINION So far, therapeutic use of activators/blockers specific for TREMs and TLTs has been limited to preclinical animal models. TREM-1 is an immediate therapeutic target for acute and chronic inflammatory conditions, especially sepsis. Certain mutations in DAP12 and TREM-2 manifest into a disorder named polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy, and newly identified TREM-2 variants confer a significant increase in risk of developing Alzheimer's disease. This makes TREM-2 an attractive therapeutic target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Christopher J Pelham
- Creighton University School of Medicine, Department of Biomedical Sciences and Center for Clinical & Translational Science , Omaha, NE 68178 , USA
| | | | | |
Collapse
|
190
|
What happens to microglial TREM2 in Alzheimer's disease: Immunoregulatory turned into immunopathogenic? Neuroscience 2014; 302:138-50. [PMID: 25281879 DOI: 10.1016/j.neuroscience.2014.09.050] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 09/23/2014] [Accepted: 09/23/2014] [Indexed: 12/12/2022]
Abstract
Microglia play major roles in initiation, coordination and execution of innate immunity in the brain. In the adult brain, these include maintenance of homeostasis, neuron and tissue repair, and eliminating infectious agents, apoptotic cells, and misfolded proteins. Some of these activities are accompanied by inflammatory reactions; and others are performed with no inflammatory effects. Under normal conditions, triggering receptor expressed on myeloid cells 2 (TREM2) belongs to the second category. It pairs with the adaptor protein DNAX-activating protein of 12kDa (DAP12) to induce phagocytosis of apoptotic neurons without inflammatory responses, and to regulate Toll-like receptor-mediated inflammatory responses, and microglial activation. Although ligands for TREM2 are largely unknown, the mitochondrial heat shock protein 60, expressed on cell surface of apoptotic neurons, is a specific ligand that activates TREM2-mediated phagocytosis by microglia. TREM2 also phagocytoses amyloid beta peptide in cultured cells. Several TREM2 mutations have been identified recently that increase the risk of Alzheimer's disease, Frontotemporal dementia, Parkinson's disease, and amyotrophic lateral sclerosis. Some of these mutations cause impaired proteolysis of full-length TREM2 at the plasma membrane to different degrees. The defects in the intramembrane cleavage result in dysfunction of phagocytosis signaling. The association of TREM2 mutations with neurodegenerative disease also calls for the understanding of the biology and pathological role of non-mutated TREM2 on human brains and microglia. This review provides a summary of current literature in TREM2 and DAP12 from several aspects, and proposes a theory that loss of TREM2 functions might contribute to the immunopathogenic role of microglia in Alzheimer's disease.
Collapse
|
191
|
Wei P, Xu L, Li CD, Sun FD, Chen L, Tan T, Luo SZ. Molecular dynamic simulation of the self-assembly of DAP12-NKG2C activating immunoreceptor complex. PLoS One 2014; 9:e105560. [PMID: 25148259 PMCID: PMC4141757 DOI: 10.1371/journal.pone.0105560] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 07/24/2014] [Indexed: 01/26/2023] Open
Abstract
The DAP12-NKG2C activating immunoreceptor complex is one of the multisubunit transmembrane protein complexes in which ligand-binding receptor chains assemble with dimeric signal-transducing modules through non-covalent associations in their transmembrane (TM) domains. In this work, both coarse grained and atomistic molecular dynamic simulation methods were applied to investigate the self-assembly dynamics of the transmembrane domains of the DAP12-NKG2C activating immunoreceptor complex. Through simulating the dynamics of DAP12-NKG2C TM heterotrimer and point mutations, we demonstrated that a five-polar-residue motif including: 2 Asps and 2 Thrs in DAP12 dimer, as well as 1 Lys in NKG2C TM plays an important role in the assembly structure of the DAP12-NKG2C TM heterotrimer. Furthermore, we provided clear evidences to exclude the possibility that another NKG2C could stably associate with the DAP12-NKG2C heterotrimer. Based on the simulation results, we proposed a revised model for the self-assembly of DAP12-NKG2C activating immunoreceptor complex, along with a plausible explanation for the association of only one NKG2C with a DAP12 dimer.
Collapse
Affiliation(s)
- Peng Wei
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China
| | - Lida Xu
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China
| | - Cheng-Dong Li
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China
| | - Fu-De Sun
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China
| | - Long Chen
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China
| | - Tianwei Tan
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China
| | - Shi-Zhong Luo
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China
- * E-mail:
| |
Collapse
|
192
|
Yoshida O, Kimura S, Dou L, Matta B, Yokota S, Stolz D, Geller D, Thomson AW. DAP12 deficiency in liver allografts results in enhanced donor DC migration, augmented effector T cell responses and abrogation of transplant tolerance. Am J Transplant 2014; 14:1791-805. [PMID: 24935196 PMCID: PMC4107008 DOI: 10.1111/ajt.12757] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/28/2014] [Accepted: 03/19/2014] [Indexed: 01/25/2023]
Abstract
Liver interstitial dendritic cells (DC) have been implicated in immune regulation and tolerance induction. We found that the transmembrane immuno-adaptor DNAX-activating protein of 12 kDa (DAP12) negatively regulated conventional liver myeloid (m) DC maturation and their in vivo migratory and T cell allostimulatory ability. Livers were transplanted from C57BL/6(H2(b) ) (B6) WT or DAP12(-/-) mice into WT C3H (H2(k) ) recipients. Donor mDC (H2-K(b+) CD11c(+) ) were quantified in spleens by flow cytometry. Anti-donor T cell reactivity was evaluated by ex vivo carboxyfluorescein diacetate succinimidyl ester-mixed leukocyte reaction and delayed-type hypersensitivity responses, while T effector and regulatory T cells were determined by flow analysis. A threefold to fourfold increase in donor-derived DC was detected in spleens of DAP12(-/-) liver recipients compared with those given WT grafts. Moreover, pro-inflammatory cytokine gene expression in the graft, interferon gamma (IFNγ) production by graft-infiltrating CD8(+) T cells and systemic levels of IFNγ were all elevated significantly in DAP12(-/-) liver recipients. DAP12(-/-) grafts also exhibited reduced incidences of CD4(+) Foxp3(+) cells and enhanced CD8(+) T cell IFNγ secretion in response to donor antigen challenge. Unlike WT grafts, DAP12(-/-) livers failed to induce tolerance and were rejected acutely. Thus, DAP12 expression in liver grafts regulates donor mDC migration to host lymphoid tissue, alloreactive T cell responses and transplant tolerance.
Collapse
Affiliation(s)
- O. Yoshida
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - S. Kimura
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - L. Dou
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA,Hepatic Surgery Center, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - B.M. Matta
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - S. Yokota
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - D.B. Stolz
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - D.A. Geller
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A. W. Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh PA,Corresponding author: Angus W. Thomson PhD DSc Starzl Transplantation Institute University of Pittsburgh School of Medicine 200 Lothrop Street, W1540 BST Pittsburgh, PA 15261
| |
Collapse
|
193
|
Ma J, Jiang T, Tan L, Yu JT. TYROBP in Alzheimer’s Disease. Mol Neurobiol 2014; 51:820-6. [DOI: 10.1007/s12035-014-8811-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Accepted: 07/11/2014] [Indexed: 10/25/2022]
|
194
|
Orbelyan GA, Tang F, Sally B, Solus J, Meresse B, Ciszewski C, Grenier JC, Barreiro LB, Lanier LL, Jabri B. Human NKG2E is expressed and forms an intracytoplasmic complex with CD94 and DAP12. THE JOURNAL OF IMMUNOLOGY 2014; 193:610-6. [PMID: 24935923 DOI: 10.4049/jimmunol.1400556] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The NKG2 family of NK receptors includes activating and inhibitory members. With the exception of the homodimer-forming NKG2D, NKG2 receptors recognize the nonclassical MHC class I molecule HLA-E, and they can be subdivided into two groups: those that associate with and signal through DAP12 to activate cells, and those that contain an ITIM motif to promote inhibition. The function of NKG2 family member NKG2E is unclear in humans, and its surface expression has never been conclusively established, largely because there is no Ab that binds specifically to NKG2E. Seeking to determine a role for this molecule, we chose to investigate its expression and ability to form complexes with intracellular signaling molecules. We found that NKG2E was capable of associating with CD94 and DAP12 but that the complex was retained intracellularly at the endoplasmic reticulum instead of being expressed on cell surfaces, and that this localization was dependent on a sequence of hydrophobic amino acids in the extracellular domain of NKG2E. Because this particular sequence has emerged and been conserved selectively among higher order primates evolutionarily, this observation raises the intriguing possibility that NKG2E may function as an intracellular protein.
Collapse
Affiliation(s)
| | - Fangming Tang
- Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Benjamin Sally
- Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Jason Solus
- Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Bertrand Meresse
- Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Cezary Ciszewski
- Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Jean-Christophe Grenier
- Sainte-Justine Hospital Research Centre, Montreal, Quebec H3T 1C5, Canada; Department of Pediatrics, University of Montreal, Montreal, Quebec H3T 1C5, Canada; and
| | - Luis B Barreiro
- Sainte-Justine Hospital Research Centre, Montreal, Quebec H3T 1C5, Canada; Department of Pediatrics, University of Montreal, Montreal, Quebec H3T 1C5, Canada; and
| | - Lewis L Lanier
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143
| | - Bana Jabri
- Department of Medicine, University of Chicago, Chicago, IL 60637;
| |
Collapse
|
195
|
Giebel S, Boratyn-Nowicka A, Karabon L, Jedynak A, Pamula-Pilat J, Tecza K, Kula D, Kowal M, Frydecka I, Grzybowska E. Associations between genes for killer immunoglobulin-like receptors and their ligands in patients with epithelial ovarian cancer. Hum Immunol 2014; 75:508-13. [DOI: 10.1016/j.humimm.2014.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 01/19/2014] [Accepted: 04/05/2014] [Indexed: 10/25/2022]
|
196
|
Ivarsson MA, Michaëlsson J, Fauriat C. Activating killer cell Ig-like receptors in health and disease. Front Immunol 2014; 5:184. [PMID: 24795726 PMCID: PMC4001058 DOI: 10.3389/fimmu.2014.00184] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 04/07/2014] [Indexed: 01/08/2023] Open
Abstract
Expression of non-rearranged HLA class I-binding receptors characterizes human and mouse NK cells. The postulation of the missing-self hypothesis some 30 years ago triggered the subsequent search and discovery of inhibitory MHC-receptors, both in humans and mice. These receptors have two functions: (i) to control the threshold for NK cell activation, a process termed “licensing” or “education,” and (ii) to inhibit NK cell activation during interactions with healthy HLA class I-expressing cells. The discovery of activating forms of KIRs (aKIR) challenged the concept of NK cell tolerance in steady state, as well as during immune challenge: what is the biological role of the activating KIR, in particular when NK cells express aKIRs in the absence of inhibitory receptors? Recently it was shown that aKIRs also participate in the education of NK cells. However, instead of lowering the threshold of activation like iKIRs, the expression of aKIRs has the opposite effect, i.e., rendering NK cells hyporesponsive. These findings may have consequences during NK cell response to viral infection, in cancer development, and in the initial stages of pregnancy. Here we review the current knowledge of activating KIRs, including the biological concept of aKIR-dependent NK cell education, and their impact in health and disease.
Collapse
Affiliation(s)
- Martin A Ivarsson
- Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge , Stockholm , Sweden
| | - Jakob Michaëlsson
- Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge , Stockholm , Sweden
| | - Cyril Fauriat
- U1068, CRCM, Immunity and Cancer, INSERM , Marseille , France ; Institut Paoli-Calmettes , Marseille , France ; UM 105, Aix-Marseille Université , Marseille , France ; UMR 7258, CNRS , Marseille , France ; U1068, CRCM, Plateforme d'Immunomonitoring en Cancérologie, INSERM , Marseille , France
| |
Collapse
|
197
|
Montalvo V, Quigley L, Vistica BP, Boelte KC, Nugent LF, Takai T, McVicar DW, Gery I. Environmental factors determine DAP12 deficiency to either enhance or suppress immunopathogenic processes. Immunology 2014; 140:475-82. [PMID: 23906311 DOI: 10.1111/imm.12158] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/28/2013] [Accepted: 07/29/2013] [Indexed: 12/30/2022] Open
Abstract
DNAX-activation protein 12 (DAP12), a transmembrane adapter, plays a major role in transducing activation signals in natural killer cells and various myeloid cells. Quantitative RT-PCR detected in normal mouse eyes considerable levels of DAP12 and multiple DAP12-coupled receptors, in particular TREM-1, Clec5a and SIRPb1. The role of DAP12 and its receptors in experimental autoimmune diseases has been controversial. Here, we analysed the effect of DAP12 deficiency on the capacity of mice to mount immunopathogenic cellular responses to the uveitogenic ocular antigen and interphotoreceptor retinoid-binding protein (IRBP), and to develop experimental autoimmune uveitis (EAU). Surprisingly, sequential analysis of EAU in mice deficient in DAP12 in two different animal facilities at first revealed enhanced disease as compared with wild-type mice, but when these mice were re-derived into a second, cleaner, animal facility, the response of control mice was essentially unchanged, whereas the DAP12 null mice were markedly hyporesponsive relative to controls in the new facility. Accordingly, when stimulated in vitro with IRBP, lymphocytes from the DAP12-deficient mice housed in the two facilities proliferated and produced opposite profiles of pro-inflammatory and anti-inflammatory cytokines, compared with their controls. These findings therefore demonstrate that the effects of DAP12 deficiency on development of autoimmune disease are dramatically affected by environmental factors.
Collapse
Affiliation(s)
- Vanessa Montalvo
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
198
|
NK Cells as a Barrier to Xenotransplantation. Xenotransplantation 2014. [DOI: 10.1128/9781555818043.ch4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
199
|
Chen X, Eksioglu EA, Zhou J, Zhang L, Djeu J, Fortenbery N, Epling-Burnette P, Van Bijnen S, Dolstra H, Cannon J, Youn JI, Donatelli SS, Qin D, De Witte T, Tao J, Wang H, Cheng P, Gabrilovich DI, List A, Wei S. Induction of myelodysplasia by myeloid-derived suppressor cells. J Clin Invest 2014; 123:4595-611. [PMID: 24216507 DOI: 10.1172/jci67580] [Citation(s) in RCA: 261] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 08/15/2013] [Indexed: 01/08/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are age-dependent stem cell malignancies that share biological features of activated adaptive immune response and ineffective hematopoiesis. Here we report that myeloid-derived suppressor cells (MDSC), which are classically linked to immunosuppression, inflammation, and cancer, were markedly expanded in the bone marrow of MDS patients and played a pathogenetic role in the development of ineffective hematopoiesis. These clonally distinct MDSC overproduce hematopoietic suppressive cytokines and function as potent apoptotic effectors targeting autologous hematopoietic progenitors. Using multiple transfected cell models, we found that MDSC expansion is driven by the interaction of the proinflammatory molecule S100A9 with CD33. These 2 proteins formed a functional ligand/receptor pair that recruited components to CD33’s immunoreceptor tyrosine-based inhibition motif (ITIM), inducing secretion of the suppressive cytokines IL-10 and TGF-β by immature myeloid cells. S100A9 transgenic mice displayed bone marrow accumulation of MDSC accompanied by development of progressive multilineage cytopenias and cytological dysplasia. Importantly, early forced maturation of MDSC by either all-trans-retinoic acid treatment or active immunoreceptor tyrosine-based activation motif–bearing (ITAM-bearing) adapter protein (DAP12) interruption of CD33 signaling rescued the hematologic phenotype. These findings indicate that primary bone marrow expansion of MDSC driven by the S100A9/CD33 pathway perturbs hematopoiesis and contributes to the development of MDS.
Collapse
|
200
|
Filtjens J, Foquet L, Taveirne S, Van Ammel E, Vanhees M, Van Acker A, Kerre T, Taghon T, Vandekerckhove B, Plum J, Van den Steen PE, Leclercq G. Contribution of the Ly49E natural killer receptor in the immune response to Plasmodium berghei infection and control of hepatic parasite development. PLoS One 2014; 9:e87463. [PMID: 24498110 PMCID: PMC3907506 DOI: 10.1371/journal.pone.0087463] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 12/28/2013] [Indexed: 12/12/2022] Open
Abstract
Natural killer (NK) cells have different roles in the host response against Plasmodium-induced malaria depending on the stage of infection. Liver NK cells have a protective role during the initial hepatic stage of infection by production of the TH1-type cytokines IFN-γ and TNF-α. In the subsequent erythrocytic stage of infection, NK cells also induce protection through Th1-type cytokines but, in addition, may also promote development of cerebral malaria via CXCR3-induction on CD8+ T cells resulting in migration of these cells to the brain. We have recently shown that the regulatory Ly49E NK receptor is expressed on liver NK cells in particular. The main objective of this study was therefore to examine the role of Ly49E expression in the immune response upon Plasmodium berghei ANKA infection, for which we compared wild type (WT) to Ly49E knockout (KO) mice. We show that the parasitemia was higher at the early stage, i.e. at days 6–7 of Plasmodium berghei ANKA infection in Ly49E KO mice, which correlated with lower induction of CD69, IFN-γ and TNF-α in DX5− liver NK cells at day 5 post-infection. At later stages, these differences faded. There was also no difference in the kinetics and the percentage of cerebral malaria development and in lymphocyte CXCR3 expression in WT versus Ly49E KO mice. Collectively, we show that the immune response against Plasmodium berghei ANKA infection is not drastically affected in Ly49E KO mice. Although NK cells play a crucial role in Plasmodium infection and Ly49E is highly expressed on liver NK cells, the Ly49E NK receptor only has a temporarily role in the immune control of this parasite.
Collapse
Affiliation(s)
- Jessica Filtjens
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium
| | - Lander Foquet
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium
| | - Sylvie Taveirne
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium
| | - Els Van Ammel
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium
| | - Mandy Vanhees
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium
| | - Aline Van Acker
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium
| | - Tessa Kerre
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium
| | - Tom Taghon
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium
| | - Bart Vandekerckhove
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium
| | - Jean Plum
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium
| | - Philippe E. Van den Steen
- Laboratory of Immunobiology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Georges Leclercq
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium
- * E-mail:
| |
Collapse
|