51
|
Mi YJ, Chen H, Guo N, Sun MY, Zhao ZH, Gao XC, Wang XL, Zhang RS, Zhou JB, Gou XC. Inhibition of PirB Activity by TAT-PEP Improves Mouse Motor Ability and Cognitive Behavior. Front Aging Neurosci 2017; 9:199. [PMID: 28676756 PMCID: PMC5476690 DOI: 10.3389/fnagi.2017.00199] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 06/02/2017] [Indexed: 01/07/2023] Open
Abstract
Paired immunoglobulin-like receptor B (PirB), a functional receptor for myelin-associated inhibitory proteins, plays an important role in axon regeneration in injured brains. However, its role in normal brain function with age has not been previously investigated. Therefore in this study, we examined the expression level of PirB in the cerebral cortex, hippocampus and cerebellum of mice at 1 month, 3 months and 18 months of age. The results showed that the expression of PirB increased with age. We further demonstrated that overexpression of PirB inhibited neurite outgrowth in PC12 cells, and this inhibitory activity of PirB could be reversed by TAT-PEP, which is a recombinant soluble PirB ectodomain fused with TAT domain for blood-brain barrier penetration. In vivo study, intraperitoneal administration of TAT-PEP was capable of enhancing motor capacity and spatial learning and memory in mice, which appeared to be mediated through regulation of brain-derived neurotrophic factor (BDNF) secretion. Our study suggests that PirB is associated with aging and TAT-PEP may be a promising therapeutic agent for modulation of age-related motor and cognitive dysfunctions.
Collapse
Affiliation(s)
- Ya-Jing Mi
- Institute of Basic and Translational Medicine, and School of Basic Medical Sciences, and Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical UniversityXi'an, China
| | - Hai Chen
- Institute of Basic and Translational Medicine, and School of Basic Medical Sciences, and Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical UniversityXi'an, China.,Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical UniversityXi'an, China
| | - Na Guo
- Institute of Basic and Translational Medicine, and School of Basic Medical Sciences, and Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical UniversityXi'an, China
| | - Meng-Yi Sun
- Department of Neurosurgery, School of Medicine, Yale UniversityNew Haven, CT, United States
| | - Zhao-Hua Zhao
- Institute of Basic and Translational Medicine, and School of Basic Medical Sciences, and Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical UniversityXi'an, China
| | - Xing-Chun Gao
- Institute of Basic and Translational Medicine, and School of Basic Medical Sciences, and Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical UniversityXi'an, China
| | - Xiao-Long Wang
- Institute of Basic and Translational Medicine, and School of Basic Medical Sciences, and Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical UniversityXi'an, China
| | - Rui-San Zhang
- Institute of Basic and Translational Medicine, and School of Basic Medical Sciences, and Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical UniversityXi'an, China
| | - Jiang-Bing Zhou
- Department of Neurosurgery, School of Medicine, Yale UniversityNew Haven, CT, United States
| | - Xing-Chun Gou
- Institute of Basic and Translational Medicine, and School of Basic Medical Sciences, and Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical UniversityXi'an, China.,Department of Neurosurgery, School of Medicine, Yale UniversityNew Haven, CT, United States
| |
Collapse
|
52
|
Ukai H, Kawahara A, Hirayama K, Case MJ, Aino S, Miyabe M, Wakita K, Oogi R, Kasayuki M, Kawashima S, Sugimoto S, Chikamatsu K, Nitta N, Koga T, Shigemoto R, Takai T, Ito I. PirB regulates asymmetries in hippocampal circuitry. PLoS One 2017; 12:e0179377. [PMID: 28594961 PMCID: PMC5464656 DOI: 10.1371/journal.pone.0179377] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 05/30/2017] [Indexed: 11/19/2022] Open
Abstract
Left-right asymmetry is a fundamental feature of higher-order brain structure; however, the molecular basis of brain asymmetry remains unclear. We recently identified structural and functional asymmetries in mouse hippocampal circuitry that result from the asymmetrical distribution of two distinct populations of pyramidal cell synapses that differ in the density of the NMDA receptor subunit GluRε2 (also known as NR2B, GRIN2B or GluN2B). By examining the synaptic distribution of ε2 subunits, we previously found that β2-microglobulin-deficient mice, which lack cell surface expression of the vast majority of major histocompatibility complex class I (MHCI) proteins, do not exhibit circuit asymmetry. In the present study, we conducted electrophysiological and anatomical analyses on the hippocampal circuitry of mice with a knockout of the paired immunoglobulin-like receptor B (PirB), an MHCI receptor. As in β2-microglobulin-deficient mice, the PirB-deficient hippocampus lacked circuit asymmetries. This finding that MHCI loss-of-function mice and PirB knockout mice have identical phenotypes suggests that MHCI signals that produce hippocampal asymmetries are transduced through PirB. Our results provide evidence for a critical role of the MHCI/PirB signaling system in the generation of asymmetries in hippocampal circuitry.
Collapse
Affiliation(s)
- Hikari Ukai
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Aiko Kawahara
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Keiko Hirayama
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Matthew Julian Case
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Shotaro Aino
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Masahiro Miyabe
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Ken Wakita
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Ryohei Oogi
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Michiyo Kasayuki
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Shihomi Kawashima
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Shunichi Sugimoto
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Kanako Chikamatsu
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Noritaka Nitta
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Tsuneyuki Koga
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Ryuichi Shigemoto
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Toshiyuki Takai
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Isao Ito
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
- * E-mail:
| |
Collapse
|
53
|
Zhao ZH, Deng B, Xu H, Zhang JF, Mi YJ, Meng XZ, Gou XC, Xu LX. PirB Overexpression Exacerbates Neuronal Apoptosis by Inhibiting TrkB and mTOR Phosphorylation After Oxygen and Glucose Deprivation Injury. Cell Mol Neurobiol 2017; 37:707-715. [PMID: 27443384 DOI: 10.1007/s10571-016-0406-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 07/08/2016] [Indexed: 11/24/2022]
Abstract
Previous studies have proven that paired immunoglobulin-like receptor B (PirB) plays a crucial suppressant role in neurite outgrowth and neuronal plasticity after central nervous system injury. However, the role of PirB in neuronal survival after cerebral ischemic injury and its mechanisms remains unclear. In the present study, the role of PirB is investigated in the survival and apoptosis of cerebral cortical neurons in cultured primary after oxygen and glucose deprivation (OGD)-induced injury. The results have shown that rebarbative PirB exacerbates early neuron apoptosis and survival. PirB gene silencing remarkably decreases early apoptosis and promotes neuronal survival after OGD. The expression of bcl-2 markedly increased and the expression of bax significantly decreased in PirB RNAi-treated neurons, as compared with the control- and control RNAi-treated ones. Further, phosphorylated TrkB and mTOR levels are significantly downregulated in the damaged neurons. However, the PirB silencing markedly upregulates phosphorylated TrkB and mTOR levels in the neurons after the OGD. Taken together, the overexpression of PirB inhibits the neuronal survival through increased neuron apoptosis. Importantly, the inhibition of the phosphorylation of TrkB and mTOR may be one of its mechanisms.
Collapse
Affiliation(s)
- Zhao-Hua Zhao
- Department of Anesthesiology, College of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
- Institute of Basic and Translational Medicine & School of Basic Medical Sciences, Xi'an Medical University, Xi'an, 710021, China
| | - Bin Deng
- Department of Anesthesiology, College of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Hao Xu
- Institute of Basic and Translational Medicine & School of Basic Medical Sciences, Xi'an Medical University, Xi'an, 710021, China
| | - Jun-Feng Zhang
- Institute of Basic and Translational Medicine & School of Basic Medical Sciences, Xi'an Medical University, Xi'an, 710021, China
| | - Ya-Jing Mi
- Institute of Basic and Translational Medicine & School of Basic Medical Sciences, Xi'an Medical University, Xi'an, 710021, China
| | - Xiang-Zhong Meng
- Department of Anesthesiology, College of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Xing-Chun Gou
- Department of Anesthesiology, College of Stomatology, Fourth Military Medical University, Xi'an, 710032, China.
- Institute of Basic and Translational Medicine & School of Basic Medical Sciences, Xi'an Medical University, Xi'an, 710021, China.
| | - Li-Xian Xu
- Department of Anesthesiology, College of Stomatology, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
54
|
Li Y, Sun Z, Cao Q, Chen M, Luo H, Lin X, Xiao F. Role of amyloid β protein receptors in mediating synaptic plasticity. Biomed Rep 2017; 6:379-386. [PMID: 28413635 PMCID: PMC5374942 DOI: 10.3892/br.2017.863] [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] [Received: 10/26/2016] [Accepted: 12/01/2016] [Indexed: 11/05/2022] Open
Abstract
There are few diseases in modern biomedicine that have garnered as much scientific interest and public concern as Alzheimer's disease (AD). The amyloid hypothesis has become the dominant model of AD pathogenesis; however, the details of the hypothesis are changing over time. Recently, given the increasing recognition, subtle effects of amyloid β protein (Aβ) on synaptic efficacy may be critical to AD progression. Synaptic plasticity is the important neurochemical foundation of learning and memory. Recent studies have identified that soluble Aβ oligomers combine with certain receptors to impair synaptic plasticity in AD, which advanced the amyloid hypothesis. The aim of the present review was to summarize the role of Aβ-relevant receptors in regulating synaptic plasticity and their downstream signaling cascades, which may provide novel insights into the understanding of the pathogenesis of AD and the development of therapeutic strategies to slow down the progression of AD-associated memory decline in the early stages.
Collapse
Affiliation(s)
- Yu Li
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Zhongqing Sun
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Qiaoyu Cao
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China
| | - Huanmin Luo
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Xi Lin
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Fei Xiao
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| |
Collapse
|
55
|
Unsworth AJ, Bye AP, Gibbins JM. Platelet-Derived Inhibitors of Platelet Activation. PLATELETS IN THROMBOTIC AND NON-THROMBOTIC DISORDERS 2017. [PMCID: PMC7123044 DOI: 10.1007/978-3-319-47462-5_37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
56
|
Kang X, Kim J, Deng M, John S, Chen H, Wu G, Phan H, Zhang CC. Inhibitory leukocyte immunoglobulin-like receptors: Immune checkpoint proteins and tumor sustaining factors. Cell Cycle 2016; 15:25-40. [PMID: 26636629 PMCID: PMC4825776 DOI: 10.1080/15384101.2015.1121324] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Inhibitory leukocyte immunoglobulin-like receptors (LILRBs 1-5) transduce signals via intracellular immunoreceptor tyrosine-based inhibitory motifs (ITIMs) that recruit protein tyrosine phosphatase non-receptor type 6 (PTPN6 or SHP-1), protein tyrosine phosphatase non-receptor type 11 (PTPN11 or SHP-2), or Src homology 2 domain-containing inositol phosphatase (SHIP), leading to negative regulation of immune cell activation. Certain of these receptors also play regulatory roles in neuronal activity and osteoclast development. The activation of LILRBs on immune cells by their ligands may contribute to immune evasion by tumors. Recent studies found that several members of LILRB family are expressed by tumor cells, notably hematopoietic cancer cells, and may directly regulate cancer development and relapse as well as the activity of cancer stem cells. LILRBs thus have dual concordant roles in tumor biology - as immune checkpoint molecules and as tumor-sustaining factors. Importantly, the study of knockout mice indicated that LILRBs do not affect hematopoiesis and normal development. Therefore LILRBs may represent ideal targets for tumor treatment. This review aims to summarize current knowledge on expression patterns, ligands, signaling, and functions of LILRB family members in the context of cancer development.
Collapse
Affiliation(s)
- Xunlei Kang
- a Department of Physiology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Jaehyup Kim
- a Department of Physiology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Mi Deng
- a Department of Physiology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Samuel John
- a Department of Physiology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Heyu Chen
- a Department of Physiology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Guojin Wu
- a Department of Physiology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Hiep Phan
- a Department of Physiology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Cheng Cheng Zhang
- a Department of Physiology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| |
Collapse
|
57
|
Bombeiro AL, Thomé R, Oliveira Nunes SL, Monteiro Moreira B, Verinaud L, de Oliveira ALR. MHC-I and PirB Upregulation in the Central and Peripheral Nervous System following Sciatic Nerve Injury. PLoS One 2016; 11:e0161463. [PMID: 27551751 PMCID: PMC4995013 DOI: 10.1371/journal.pone.0161463] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 08/05/2016] [Indexed: 12/12/2022] Open
Abstract
Major histocompatibility complex class one (MHC-I) antigen-presenting molecules participate in central nervous system (CNS) synaptic plasticity, as does the paired immunoglobulin-like receptor B (PirB), an MHC-I ligand that can inhibit immune-cells and bind to myelin axon growth inhibitors. Based on the dual roles of both molecules in the immune and nervous systems, we evaluated their expression in the central and peripheral nervous system (PNS) following sciatic nerve injury in mice. Increased PirB and MHC-I protein and gene expression is present in the spinal cord one week after nerve transection, PirB being mostly expressed in the neuropile region. In the crushed nerve, MHC-I protein levels increased 2 weeks after lesion (wal) and progressively decreased over the next eight weeks. The same kinetics were observed for infiltrating cytotoxic T lymphocytes (CTLs) but not for PirB expression, which continuously increased. Both MHC-I and PirB were found in macrophages and Schwann cells but rarely in axons. Interestingly, at 8 wal, PirB was mainly restricted to the myelin sheath. Our findings reinforce the participation of MHC-I and PirB in CNS plasticity events. In contrast, opposing expression levels of these molecules were found in the PNS, so that MHC-I and PirB seem to be mostly implicated in antigen presentation to CTLs and axon myelination, respectively.
Collapse
Affiliation(s)
- André Luis Bombeiro
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas – UNICAMP, Rua Monteiro Lobato, 255, CEP: 13083–865, Campinas, SP, Brazil
| | - Rodolfo Thomé
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas – UNICAMP, Rua Monteiro Lobato, 255, CEP: 13083–865, Campinas, SP, Brazil
| | - Sérgio Luiz Oliveira Nunes
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas – UNICAMP, Rua Monteiro Lobato, 255, CEP: 13083–865, Campinas, SP, Brazil
| | - Bárbara Monteiro Moreira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas – UNICAMP, Rua Monteiro Lobato, 255, CEP: 13083–865, Campinas, SP, Brazil
| | - Liana Verinaud
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas – UNICAMP, Rua Monteiro Lobato, 255, CEP: 13083–865, Campinas, SP, Brazil
| | - Alexandre Leite Rodrigues de Oliveira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas – UNICAMP, Rua Monteiro Lobato, 255, CEP: 13083–865, Campinas, SP, Brazil
- * E-mail:
| |
Collapse
|
58
|
von Websky MW, Kitamura K, Ludwig-Portugall I, Kurts C, von Laffert M, LeMaoult J, Carosella ED, Abu-Elmagd K, Kalff JC, Schäfer N. Recombinant HLA-G as Tolerogenic Immunomodulant in Experimental Small Bowel Transplantation. PLoS One 2016; 11:e0158907. [PMID: 27404095 PMCID: PMC4942037 DOI: 10.1371/journal.pone.0158907] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/23/2016] [Indexed: 11/18/2022] Open
Abstract
The non-classical MHC I paralogue HLA-G is expressed by cytotrophoblast cells and implicated with fetomaternal tolerance by downregulating the maternal adaptive and innate immune response against the fetus. HLA-G expression correlates with favorable graft outcome in humans and recently promising immunosuppressive effects of therapeutic HLA-G in experimental transplantation (skin allograft acceptance) were shown. Consequently, we examined this novel therapeutic approach in solid organ transplantation. In this study, therapeutic recombinant HLA-G5 was evaluated for the first time in a solid organ model of acute rejection (ACR) after orthotopic intestinal transplantation (ITX). Allogenic ITX was performed in rats (Brown Norway to Lewis) with and without HLA-G treatment. It was found that HLA-G treatment significantly reduced histologically proven ACR at both an early and late postoperative timepoint (POD 4/7), concomitant to a functionally preserved graft contractility at POD 7. Interestingly, graft infiltration by myeloperoxidase+ cells was significantly reduced at POD7 by HLA-G treatment. Moreover, HLA-G treatment showed an effect on the allogenic T-cell immune response as assessed by flow cytometry: The influx of recipient-derived CD8+ T-cells into the graft mesenteric lymphnodes at POD7 was significantly reduced while CD4+ populations were not affected. As a potential mechanism of action, an induction of T-reg populations in the mesenteric lymphnodes was postulated, but flow cytometric analysis of classical CD4+/CD25+/FoxP3+Treg-cells showed no significant alteration by HLA-G treatment. The novel therapeutic approach using recombinant HLA-G5 reported herein demonstrates a significant immunosuppressive effect in this model of allogenic experimental intestinal transplantation. This effect may be mediated via inhibition of recipient-derived CD8+ T-cell populations either directly or by induction of non-classical Treg populations.
Collapse
Affiliation(s)
| | - Koji Kitamura
- Department of Surgery, University Hospital of Bonn, Bonn, Germany
- Department of Hepatobiliary Pancreatic Surgery and Transplantation, Kyoto University Hospital, Kyoto, Japan
| | | | - Christian Kurts
- Institute for Experimental Immunology, University of Bonn, Germany
| | | | - Joel LeMaoult
- CEA, iMETI, Research Division in Hematology and Immunology, Saint-Louis Hospital, Paris, France
| | - Edgardo D. Carosella
- CEA, iMETI, Research Division in Hematology and Immunology, Saint-Louis Hospital, Paris, France
| | - Kareem Abu-Elmagd
- Transplant Center, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Joerg C. Kalff
- Department of Surgery, University Hospital of Bonn, Bonn, Germany
| | - Nico Schäfer
- Department of Surgery, University Hospital of Bonn, Bonn, Germany
- * E-mail:
| |
Collapse
|
59
|
Wakefield D, Yates W, Amjadi S, McCluskey P. HLA-B27 Anterior Uveitis: Immunology and Immunopathology. Ocul Immunol Inflamm 2016; 24:450-9. [PMID: 27245590 DOI: 10.3109/09273948.2016.1158283] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Acute anterior uveitis (AAU) is the commonest type of uveitis and HLA-B27 AAU is the most frequently recognized type of acute anterior uveitis and anterior uveitis overall. Recent evidence indicates that acute anterior uveitis is a heterogenous disease, is polygenic and is frequently associated with the spondyloarthropathies (SpA). Studies of patients with AAU and animal models of disease indicate a role for innate immunity, the IL-23 cytokine pathway and exogenous factors, in the pathogenesis of both SpA and acute anterior uveitis. Recently described genetic associations cluster around immunologic pathways, including the IL-17 and IL-23 pathways, antigen processing and presentation, and lymphocyte development and activation. Patients with ankylosing spondylitis (AS) and AAU share other genetic markers, such as ERAP-1, which show strong evidence of gene-gene interaction and point to new mechanisms of disease pathogenesis. These observations have major implications for understanding the pathogenesis of HLA-B27 diseases, such as AAU, and may lead to the development of more specific therapy for AAU. Received 6 January 2016; revised 6 February 2016; accepted 18 February 2016; published online 31 May 2016.
Collapse
Affiliation(s)
- Denis Wakefield
- a Laboratory of Ocular Immunology , University of New South Wales , Kensington , Sydney , Australia
| | - William Yates
- a Laboratory of Ocular Immunology , University of New South Wales , Kensington , Sydney , Australia
| | - Shahriar Amjadi
- a Laboratory of Ocular Immunology , University of New South Wales , Kensington , Sydney , Australia
| | - Peter McCluskey
- b Save Sight Institute, Discipline of Ophthalmology , Sydney Medical School, The University of Sydney , Sydney , Australia
| |
Collapse
|
60
|
Microbially cleaved immunoglobulins are sensed by the innate immune receptor LILRA2. Nat Microbiol 2016; 1:16054. [DOI: 10.1038/nmicrobiol.2016.54] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 03/22/2016] [Indexed: 12/15/2022]
|
61
|
van Rees DJ, Szilagyi K, Kuijpers TW, Matlung HL, van den Berg TK. Immunoreceptors on neutrophils. Semin Immunol 2016; 28:94-108. [PMID: 26976825 PMCID: PMC7129252 DOI: 10.1016/j.smim.2016.02.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 02/24/2016] [Accepted: 02/26/2016] [Indexed: 12/12/2022]
Abstract
Neutrophil activities must be tightly controlled to maintain immune homeostasis. Activating and inhibitory receptors balance the outcome of immune cell activation. Immunoreceptors contain Ig-like extracellular domains and signal via ITAMs or ITIMs. Syk or SHP/SHIP mediate downstream signaling after immunoreceptor activation. Targeting immunoreceptors provides opportunities for therapeutic interventions.
Neutrophils play a critical role in the host defense against infection, and they are able to perform a variety of effector mechanisms for this purpose. However, there are also a number of pathological conditions, including autoimmunity and cancer, in which the activities of neutrophils can be harmful to the host. Thus the activities of neutrophils need to be tightly controlled. As in the case of other immune cells, many of the neutrophil effector functions are regulated by a series of immunoreceptors on the plasma membrane. Here, we review what is currently known about the functions of the various individual immunoreceptors and their signaling in neutrophils. While these immunoreceptors allow for the recognition of a diverse range of extracellular ligands, such as cell surface structures (like proteins, glycans and lipids) and extracellular matrix components, they commonly signal via conserved ITAM or ITIM motifs and their associated downstream pathways that depend on the phosphorylation of tyrosine residues in proteins and/or inositol lipids. This allows for a balanced homeostatic regulation of neutrophil effector functions. Given the number of available immunoreceptors and their fundamental importance for neutrophil behavior, it is perhaps not surprising that pathogens have evolved means to evade immune responses through some of these pathways. Inversely, some of these receptors evolved to specifically recognize these pathogens. Finally, some interactions mediated by immunoreceptors in neutrophils have been identified as promising targets for therapeutic intervention.
Collapse
Affiliation(s)
- Dieke J van Rees
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Katka Szilagyi
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Taco W Kuijpers
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Hanke L Matlung
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Timo K van den Berg
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
62
|
Metabolism Is Central to Tolerogenic Dendritic Cell Function. Mediators Inflamm 2016; 2016:2636701. [PMID: 26980944 PMCID: PMC4766347 DOI: 10.1155/2016/2636701] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 12/31/2015] [Indexed: 12/13/2022] Open
Abstract
Immunological tolerance is a fundamental tenant of immune homeostasis and overall health. Self-tolerance is a critical component of the immune system that allows for the recognition of self, resulting in hyporeactivity instead of immunogenicity. Dendritic cells are central to the establishment of dominant immune tolerance through the secretion of immunosuppressive cytokines and regulatory polarization of T cells. Cellular metabolism holds the key to determining DC immunogenic or tolerogenic cell fate. Recent studies have demonstrated that dendritic cell maturation leads to a shift toward a glycolytic metabolic state and preferred use of glucose as a carbon source. In contrast, tolerogenic dendritic cells favor oxidative phosphorylation and fatty acid oxidation. This dichotomous metabolic reprogramming of dendritic cells drives differential cellular function and plays a role in pathologies, such as autoimmune disease. Pharmacological alterations in metabolism have promising therapeutic potential.
Collapse
|
63
|
Han SH, Park JC, Mook-Jung I. Amyloid β-interacting partners in Alzheimer's disease: From accomplices to possible therapeutic targets. Prog Neurobiol 2016; 137:17-38. [DOI: 10.1016/j.pneurobio.2015.12.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 12/02/2015] [Accepted: 12/09/2015] [Indexed: 12/20/2022]
|
64
|
Hofer J, Forster F, Isenman DE, Wahrmann M, Leitner J, Hölzl MA, Kovarik JJ, Stockinger H, Böhmig GA, Steinberger P, Zlabinger GJ. Ig-like transcript 4 as a cellular receptor for soluble complement fragment C4d. FASEB J 2015; 30:1492-503. [PMID: 26678451 DOI: 10.1096/fj.15-275594] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 12/08/2015] [Indexed: 01/16/2023]
Abstract
Complement regulation leads to the generation of complement split products (CSPs) such as complement component (C)4d, a marker for disease activity in autoimmune syndromes or antibody-mediated allograft rejection. However, the physiologic role of C4d has been unknown. By screening murine thymoma BW5147 cells expressing a cDNA library generated from human monocyte-derived dendritic cells with recombinant human C4d, we identified Ig-like transcript (ILT)4 and ILT5v2 as cellular receptors for C4d. Both receptors, expressed on monocytes, macrophages, and dendritic cells, also interacted with the CSPs C3d, C4b, C3b, and iC3b. However, C4d did not bind to classic complement receptors (CRs). Interaction between cell surface-resident ILT4 and soluble monomeric C4d resulted in endocytosis of C4d. Surprisingly, binding of soluble ILT4 to C4d covalently immobilized to a cellular surface following classic complement activation could not be detected. Remarkably, C4d immobilized to a solid phaseviaits intrinsic thioester conferred a dose-dependent inhibition of TNF-α and IL-6 secretion in monocytes activatedviaFc-cross-linking of up to 50% as compared to baseline. Similarly, C4d conferred an attenuation of intracellular Ca(2+)flux in monocytes activatedviaFc-cross-linking. In conclusion, ILT4 represents a scavenger-type endocytotic CR for soluble monomeric C4d, whereas attenuation of monocyte activation by physiologically oriented C4d on a surface appears to be dependent on a yet to be identified C4d receptor.-Hofer, J., Forster, F., Isenman, D. E., Wahrmann, M., Leitner, J., Hölzl, M. A., Kovarik, J. K., Stockinger, H., Böhmig, G. A., Steinberger, P., Zlabinger, G. J. Ig-like transcript 4 as a cellular receptor for soluble complement fragment C4d.
Collapse
Affiliation(s)
- Johannes Hofer
- *Division of Clinical Experimental Immunology and Division of Immune Receptors and T Cell Activation, Institute of Immunology, Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Centre for Pathophysiology, Infectiology, and Immunology, and Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; and Departments of Biochemistry and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Florian Forster
- *Division of Clinical Experimental Immunology and Division of Immune Receptors and T Cell Activation, Institute of Immunology, Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Centre for Pathophysiology, Infectiology, and Immunology, and Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; and Departments of Biochemistry and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - David E Isenman
- *Division of Clinical Experimental Immunology and Division of Immune Receptors and T Cell Activation, Institute of Immunology, Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Centre for Pathophysiology, Infectiology, and Immunology, and Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; and Departments of Biochemistry and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Markus Wahrmann
- *Division of Clinical Experimental Immunology and Division of Immune Receptors and T Cell Activation, Institute of Immunology, Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Centre for Pathophysiology, Infectiology, and Immunology, and Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; and Departments of Biochemistry and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Judith Leitner
- *Division of Clinical Experimental Immunology and Division of Immune Receptors and T Cell Activation, Institute of Immunology, Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Centre for Pathophysiology, Infectiology, and Immunology, and Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; and Departments of Biochemistry and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Markus A Hölzl
- *Division of Clinical Experimental Immunology and Division of Immune Receptors and T Cell Activation, Institute of Immunology, Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Centre for Pathophysiology, Infectiology, and Immunology, and Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; and Departments of Biochemistry and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Johannes J Kovarik
- *Division of Clinical Experimental Immunology and Division of Immune Receptors and T Cell Activation, Institute of Immunology, Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Centre for Pathophysiology, Infectiology, and Immunology, and Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; and Departments of Biochemistry and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Hannes Stockinger
- *Division of Clinical Experimental Immunology and Division of Immune Receptors and T Cell Activation, Institute of Immunology, Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Centre for Pathophysiology, Infectiology, and Immunology, and Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; and Departments of Biochemistry and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Georg A Böhmig
- *Division of Clinical Experimental Immunology and Division of Immune Receptors and T Cell Activation, Institute of Immunology, Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Centre for Pathophysiology, Infectiology, and Immunology, and Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; and Departments of Biochemistry and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Peter Steinberger
- *Division of Clinical Experimental Immunology and Division of Immune Receptors and T Cell Activation, Institute of Immunology, Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Centre for Pathophysiology, Infectiology, and Immunology, and Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; and Departments of Biochemistry and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Gerhard J Zlabinger
- *Division of Clinical Experimental Immunology and Division of Immune Receptors and T Cell Activation, Institute of Immunology, Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Centre for Pathophysiology, Infectiology, and Immunology, and Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; and Departments of Biochemistry and Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
65
|
van Beek L, Vroegrijk IOCM, Katiraei S, Heemskerk MM, van Dam AD, Kooijman S, Rensen PCN, Koning F, Verbeek JS, Willems van Dijk K, van Harmelen V. FcRγ-chain deficiency reduces the development of diet-induced obesity. Obesity (Silver Spring) 2015; 23:2435-44. [PMID: 26523352 DOI: 10.1002/oby.21309] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/16/2015] [Accepted: 07/30/2015] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Pathogenic immunoglobulins are produced during the development of obesity and contribute to the development of insulin resistance (IR). However, the mechanisms by which these antibodies affect IR are largely unknown. This study investigated whether Fc-receptors contribute to the development of diet-induced obesity and IR by studying FcRγ(-/-) mice that lack the γ-subunit necessary for signaling and cell surface expression of FcγR and FcεRI. METHODS FcRγ(-/-) and wild-type (WT) mice were fed a high-fat diet (HFD) to induce obesity. At 4 and 11 weeks, body weight and insulin sensitivity were measured, and adipose tissue (AT) inflammation was determined. Furthermore, intestinal triglyceride (TG) uptake and plasma TG clearance were determined, and gut microbiota composition was analyzed. RESULTS FcRγ(-/-) mice gained less weight after 11 weeks of HFD. They had reduced adiposity, adipose tissue inflammation, and IR. Interestingly, FcRγ(-/-) mice had higher lean mass compared to WT mice, which was associated with increased energy expenditure. Intestinal TG absorption was increased whereas plasma TG clearance was not affected in FcRγ(-/-) mice. Gut microbial composition differed significantly and might therefore have added to the observed phenotype. CONCLUSIONS FcRγ-chain deficiency reduces the development of diet-induced obesity, as well as associated AT inflammation and IR at 11 weeks of HFD.
Collapse
Affiliation(s)
- Lianne van Beek
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Irene O C M Vroegrijk
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Department of Medicine, Division Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| | - Saeed Katiraei
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Mattijs M Heemskerk
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Andrea D van Dam
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Department of Medicine, Division Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| | - Sander Kooijman
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Department of Medicine, Division Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| | - Patrick C N Rensen
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Department of Medicine, Division Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| | - Frits Koning
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - J Sjef Verbeek
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Department of Medicine, Division Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| | - Vanessa van Harmelen
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
66
|
Liu J, Wang Y, Fu W. Axon regeneration impediment: the role of paired immunoglobulin-like receptor B. Neural Regen Res 2015; 10:1338-42. [PMID: 26487866 PMCID: PMC4590251 DOI: 10.4103/1673-5374.162771] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Regenerative capacity is weak after central nervous system injury because of the absence of an enhancing microenvironment and presence of an inhibitory microenvironment for neuronal and axonal repair. In addition to the Nogo receptor (NgR), the paired immunoglobulin-like receptor B (PirB) is a recently discovered coreceptor of Nogo, myelin-associated glycoprotein, and myelin oligodendrocyte glycoprotein. Concurrent blocking of NgR and PirB almost completely eliminates the inhibitory effect of myelin-associated inhibitory molecules on axonal regeneration. PirB participates in a key pathological process of the nervous system, specifically axonal regeneration inhibition. PirB is an inhibitory receptor similar to NgR, but their effects are not identical. This study summarizes the structure, distribution, relationship with common nervous system diseases, and known mechanisms of PirB, and concludes that PirB is also distributed in cells of the immune and hematopoietic systems. Further investigations are needed to determine if immunomodulation and blood cell migration involve inhibition of axonal regeneration.
Collapse
Affiliation(s)
- Jing Liu
- Neonatal Intensive Care Center, BAYI Children's Hospital, Beijing Military General Hospital of Chinese PLA, Beijing, China
- Correspondence to: Jing Liu,
| | - Yan Wang
- Neonatal Intensive Care Center, BAYI Children's Hospital, Beijing Military General Hospital of Chinese PLA, Beijing, China
- Graduate School, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Wei Fu
- Neonatal Intensive Care Center, BAYI Children's Hospital, Beijing Military General Hospital of Chinese PLA, Beijing, China
- Graduate School, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
67
|
Hirayasu K, Arase H. Functional and genetic diversity of leukocyte immunoglobulin-like receptor and implication for disease associations. J Hum Genet 2015; 60:703-8. [PMID: 26040207 DOI: 10.1038/jhg.2015.64] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 05/08/2015] [Accepted: 05/10/2015] [Indexed: 01/03/2023]
Abstract
Human leukocyte immunoglobulin-like receptors (LILR) are a family of 11 functional genes encoding five activating (LILRA1, 2, 4-6), five inhibitory (LILRB1-5) and one soluble (LILRA3) form. The number of LILR genes is conserved among individuals, except for LILRA3 and LILRA6, which exhibit copy-number variations. The LILR genes are rapidly evolving and showing large interspecies differences, making it difficult to analyze the functions of LILR using an animal model. LILRs are expressed on various cells such as lymphoid and myeloid cells and the expression patterns are different from gene to gene. The LILR gene expression and polymorphisms have been reported to be associated with autoimmune and infectious diseases such as rheumatoid arthritis and cytomegalovirus infection. Although human leukocyte antigen (HLA) class I is a well-characterized ligand for some LILRs, non-HLA ligands have been increasingly identified in recent years. LILRs have diverse functions, including the regulation of inflammation, immune tolerance, cell differentiation and nervous system plasticity. This review focuses on the genetic and functional diversity of the LILR family.
Collapse
Affiliation(s)
- Kouyuki Hirayasu
- Laboratory of Immunochemistry, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Hisashi Arase
- Laboratory of Immunochemistry, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| |
Collapse
|
68
|
Kishida K, Kohyama M, Kurashima Y, Kogure Y, Wang J, Hirayasu K, Suenaga T, Kiyono H, Kunisawa J, Arase H. Negative regulation of DSS-induced experimental colitis by PILRα. Int Immunol 2015; 27:307-14. [DOI: 10.1093/intimm/dxv004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 01/23/2015] [Indexed: 12/16/2022] Open
|
69
|
Ramasamy S, Yu F, Hong Yu Y, Srivats H, Dawe GS, Ahmed S. NogoR1 and PirB signaling stimulates neural stem cell survival and proliferation. Stem Cells 2015; 32:1636-48. [PMID: 24449409 DOI: 10.1002/stem.1645] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 12/11/2013] [Indexed: 11/07/2022]
Abstract
Neural stem cells (NSCs) and neural progenitors (NPs) in the mammalian neocortex give rise to the main cell types of the nervous system. The biological behavior of these NSCs and NPs is regulated by extracellular niche derived autocrine-paracrine signaling factors on a developmental timeline. Our previous reports [Plos One 2010;5:e15341; J Neurochem 2011;117:565-578] have shown that chondroitin sulfate proteoglycan and ApolipoproteinE are autocrine-paracrine survival factors for NSCs. NogoA, a myelin related protein, is expressed in the cortical ventricular zones where NSCs reside. However, the functional role of Nogo signaling proteins in NSC behavior is not completely understood. In this study, we show that NogoA receptors, NogoR1 and PirB, are expressed in the ventricular zone where NSCs reside between E10.5 and 14.5 but not at E15.5. Nogo ligands stimulate NSC survival and proliferation in a dosage-dependent manner in vitro. NogoR1 and PirB are low and high affinity Nogo receptors, respectively and are responsible for the effects of Nogo ligands on NSC behavior. Inhibition of autocrine-paracrine Nogo signaling blocks NSC survival and proliferation. In NSCs, NogoR1 functions through Rho whereas PirB uses Shp1/2 signaling pathways to control NSC behavior. Taken together, this work suggests that Nogo signaling is an important pathway for survival of NSCs.
Collapse
Affiliation(s)
- Srinivas Ramasamy
- Institute of Medical Biology, 8A Biomedical Grove, #05-37 Immunos, Singapore
| | | | | | | | | | | |
Collapse
|
70
|
The Binding Receptors of Aβ: an Alternative Therapeutic Target for Alzheimer's Disease. Mol Neurobiol 2014; 53:455-471. [PMID: 25465238 DOI: 10.1007/s12035-014-8994-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 11/06/2014] [Indexed: 01/18/2023]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative disorders, which causes the deterioration of memory and other cognitive abilities of the elderly. Previous lines of research have shown that Aβ is an essential factor in AD pathology and the soluble oligomeric species of Aβ peptide is presumed to be the drivers of synaptic impairment in AD. However, the exact mechanisms underlying Aβ-induced synapse dysfunction are still not fully understood. Recently, increasing evidence suggests that some potential receptors which bind specifically with Aβ may play important roles in inducing the toxicity of the neurons in AD pathology. These receptors include the cellular prion protein (PrPc), the α7 nicotinic acetylcholine receptor (α7nAChR), the p75 neurotrophin receptor (p75(NTR)), the beta-adrenergic receptors (β-ARs), the Eph receptors, the paired immunoglobulin-like receptor B (PirB), the PirB's human ortholog receptor (LilrB2), and the Fcγ receptor II-b (FcγRIIb). This review summarizes the characters of these prominent receptors and how the bindings of them with Aβ inhibit the LTP, decrease the number of dendritic spine, damage the neurons, and so on in AD pathogenesis. Blocking or rescuing these receptors may have significant importance for AD treatments.
Collapse
|
71
|
Fujita Y, Yamashita T. Axon growth inhibition by RhoA/ROCK in the central nervous system. Front Neurosci 2014; 8:338. [PMID: 25374504 PMCID: PMC4205828 DOI: 10.3389/fnins.2014.00338] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 10/06/2014] [Indexed: 12/31/2022] Open
Abstract
Rho kinase (ROCK) is a serine/threonine kinase and a downstream target of the small GTPase Rho. The RhoA/ROCK pathway is associated with various neuronal functions such as migration, dendrite development, and axonal extension. Evidence from animal studies reveals that RhoA/ROCK signaling is involved in various central nervous system (CNS) diseases, including optic nerve and spinal cord injuries, stroke, and neurodegenerative diseases. Given that RhoA/ROCK plays a critical role in the pathophysiology of CNS diseases, the development of therapeutic agents targeting this pathway is expected to contribute to the treatment of CNS diseases. The RhoA/ROCK pathway mediates the effects of myelin-associated axon growth inhibitors—Nogo, myelin-associated glycoprotein (MAG), oligodendrocyte-myelin glycoprotein (OMgp), and repulsive guidance molecule (RGM). Blocking RhoA/ROCK signaling can reverse the inhibitory effects of these molecules on axon outgrowth, and promotes axonal sprouting and functional recovery in animal models of CNS injury. To date, several RhoA/ROCK inhibitors have been under development or in clinical trials as therapeutic agents for neurological disorders. In this review, we focus on the RhoA/ROCK signaling pathway in neurological disorders. We also discuss the potential therapeutic approaches of RhoA/ROCK inhibitors for various neurological disorders.
Collapse
Affiliation(s)
- Yuki Fujita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University Osaka, Japan ; Japan Science and Technology Agency, Core Research for Evolutional Science and Technology Tokyo, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University Osaka, Japan ; Japan Science and Technology Agency, Core Research for Evolutional Science and Technology Tokyo, Japan
| |
Collapse
|
72
|
Lang BT, Wang J, Filous AR, Au NPB, Ma CHE, Shen Y. Pleiotropic molecules in axon regeneration and neuroinflammation. Exp Neurol 2014; 258:17-23. [DOI: 10.1016/j.expneurol.2014.04.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 04/21/2014] [Accepted: 04/29/2014] [Indexed: 12/20/2022]
|
73
|
Abstract
Murine paired immunoglobulin-like receptors B (PIRB), as the ortholog of human leukocyte immunoglobulin-like receptor B2 (LILRB2), is involved in a variety of biological functions. Here, we found that PIRB and LILRB2 were expressed in mouse and human platelets, respectively. PIRB intracellular domain deletion (PIRB-TM) mice had thrombocythemia and significantly higher proportions of megakaryocytes in bone marrow. Agonist-induced aggregation and spreading on immobilized fibrinogen were facilitated in PIRB-TM platelets. The rate of clot retraction in platelet-rich plasma containing PIRB-TM platelets was also increased. Characterization of signaling confirmed that PIRB associated with phosphatases Shp1/2 in platelets. The phosphorylation of Shp1/2 was significantly downregulated in PIRB-TM platelets stimulated with collagen-related peptide (CRP) or on spreading. The results further revealed that the phosphorylation levels of the linker for activation of T cells, SH2 domain-containing leukocyte protein of 76kDa, and phospholipase C were enhanced in PIRB-TM platelets stimulated with CRP. The phosphorylation levels of FAK Y397 and integrin β3 Y759 were also enhanced in PIRB-TM platelet spread on fibrinogen. The PIRB/LILRB2 ligand angiopoietin-like-protein 2 (ANGPTL2) was expressed and stored in platelet α-granules. ANGPTL2 inhibited agonist-induced platelet aggregation and spreading on fibrinogen. The data presented here reveal that PIRB and its ligand ANGPTL2 possess an antithrombotic function by suppressing collagen receptor glycoprotein VI and integrin αIIbβ3-mediated signaling.
Collapse
|
74
|
Murakami Y, Tian L, Voss OH, Margulies DH, Krzewski K, Coligan JE. CD300b regulates the phagocytosis of apoptotic cells via phosphatidylserine recognition. Cell Death Differ 2014; 21:1746-57. [PMID: 25034781 DOI: 10.1038/cdd.2014.86] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/14/2014] [Accepted: 05/21/2014] [Indexed: 01/15/2023] Open
Abstract
The CD300 receptor family members are a group of molecules that modulate a variety of immune cell processes. We show that mouse CD300b (CLM7/LMIR5), expressed on myeloid cells, recognizes outer membrane-exposed phosphatidylserine (PS) and does not, as previously reported, directly recognize TIM1 or TIM4. CD300b accumulates in phagocytic cups along with F-actin at apoptotic cell contacts, thereby facilitating their engulfment. The CD300b-mediated activation signal is conveyed through CD300b association with the adaptor molecule DAP12, and requires a functional DAP12 ITAM motif. Binding of apoptotic cells promotes the activation of the PI3K-Akt kinase pathway in macrophages, while silencing of CD300b expression diminishes PI3K-Akt kinase activation and impairs efferocytosis. Collectively, our data show that CD300b recognizes PS as a ligand, and regulates the phagocytosis of apoptotic cells via the DAP12 signaling pathway.
Collapse
Affiliation(s)
- Y Murakami
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - L Tian
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - O H Voss
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - D H Margulies
- Molecular Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - K Krzewski
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - J E Coligan
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
75
|
Gou Z, Mi Y, Jiang F, Deng B, Yang J, Gou X. PirB is a novel potential therapeutic target for enhancing axonal regeneration and synaptic plasticity following CNS injury in mammals. J Drug Target 2014; 22:365-71. [PMID: 24405091 DOI: 10.3109/1061186x.2013.878939] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
A major barrier to axonal regeneration in mammals is the unfavorable extracellular environment that develops following injury to the central nervous system (CNS). In particular, three myelin-associated inhibitory proteins (MAIs) - Nogo, myelin-associated glycoprotein (MAG) and oligodendrocyte myelin glycoprotein (OMgp) - are known to inhibit axonal regeneration and functional recovery. These MAIs share a common receptor, glycosylphosphatidylinositol-anchored Nogo receptor (NgR). However, paired immunoglobulin-like receptor B (PirB) - which was originally identified as a receptor for class I major histocompatibility complex (MHCI) in the immune system - is also expressed in neurones and plays a similarly inhibitory role in axonal regeneration and synaptic plasticity following CNS injury through its association with MAIs. Importantly, suppression of PirB activity through antibody antagonism or genetic means can partially relieve the inhibition of neurite outgrowth in vitro and in vivo. In this review, we present the molecular features, expression patterns and known signaling pathways of PirB, and we specifically focus on putative roles for PirB in the CNS and its potential as a target of molecular therapies for enhancing axonal regeneration and synaptic plasticity following CNS injury.
Collapse
Affiliation(s)
- Zhaoyu Gou
- College of Life Science, China West Normal University , Nanchong , China and
| | | | | | | | | | | |
Collapse
|
76
|
Paired immunoglobulin-like receptor A is an intrinsic, self-limiting suppressor of IL-5-induced eosinophil development. Nat Immunol 2013; 15:36-44. [PMID: 24212998 PMCID: PMC3869881 DOI: 10.1038/ni.2757] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 09/24/2013] [Indexed: 12/16/2022]
Abstract
Eosinophilia is a hallmark characteristic of T helper type 2 (TH2) cell-associated diseases and is critically regulated by the central eosinophil growth factor interleukin 5 (IL-5). Here we demonstrate that IL-5 activity in eosinophils was regulated by paired immunoglobulin-like receptors PIR-A and PIR-B. Upon self-recognition of β₂-microglobulin (β₂M) molecules, PIR-B served as a permissive checkpoint for IL-5-induced development of eosinophils by suppressing the proapoptotic activities of PIR-A, which were mediated by the Grb2-Erk-Bim pathway. PIR-B-deficient bone marrow eosinophils underwent compartmentalized apoptosis, resulting in decreased blood eosinophilia in naive mice and in mice challenged with IL-5. Subsequently, Pirb(-/-) mice displayed impaired aeroallergen-induced lung eosinophilia and induction of lung TH2 cell responses. Collectively, these data uncover an intrinsic, self-limiting pathway regulating IL-5-induced expansion of eosinophils, which has broad implications for eosinophil-associated diseases.
Collapse
|
77
|
Kim T, Vidal GS, Djurisic M, William CM, Birnbaum ME, Garcia KC, Hyman BT, Shatz CJ. Human LilrB2 is a β-amyloid receptor and its murine homolog PirB regulates synaptic plasticity in an Alzheimer's model. Science 2013; 341:1399-404. [PMID: 24052308 PMCID: PMC3853120 DOI: 10.1126/science.1242077] [Citation(s) in RCA: 311] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Soluble β-amyloid (Aβ) oligomers impair synaptic plasticity and cause synaptic loss associated with Alzheimer's disease (AD). We report that murine PirB (paired immunoglobulin-like receptor B) and its human ortholog LilrB2 (leukocyte immunoglobulin-like receptor B2), present in human brain, are receptors for Aβ oligomers, with nanomolar affinity. The first two extracellular immunoglobulin (Ig) domains of PirB and LilrB2 mediate this interaction, leading to enhanced cofilin signaling, also seen in human AD brains. In mice, the deleterious effect of Aβ oligomers on hippocampal long-term potentiation required PirB, and in a transgenic model of AD, PirB not only contributed to memory deficits present in adult mice, but also mediated loss of synaptic plasticity in juvenile visual cortex. These findings imply that LilrB2 contributes to human AD neuropathology and suggest therapeutic uses of blocking LilrB2 function.
Collapse
Affiliation(s)
- Taeho Kim
- Departments of Biology and Neurobiology and Bio-X, James H. Clark Center, Stanford University, Stanford, CA 94305, USA
| | - George S. Vidal
- Departments of Biology and Neurobiology and Bio-X, James H. Clark Center, Stanford University, Stanford, CA 94305, USA
| | - Maja Djurisic
- Departments of Biology and Neurobiology and Bio-X, James H. Clark Center, Stanford University, Stanford, CA 94305, USA
| | | | - Michael E. Birnbaum
- Howard Hughes Medical Institute, Department of Molecular and Cellular Physiology, and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - K. Christopher Garcia
- Howard Hughes Medical Institute, Department of Molecular and Cellular Physiology, and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bradley T. Hyman
- Neuropathology Service, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Carla J. Shatz
- Departments of Biology and Neurobiology and Bio-X, James H. Clark Center, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
78
|
|
79
|
Scroggins SM, Olivier AK, Meyerholz DK, Schlueter AJ. Characterization of regulatory dendritic cells that mitigate acute graft-versus-host disease in older mice following allogeneic bone marrow transplantation. PLoS One 2013; 8:e75158. [PMID: 24040397 PMCID: PMC3769303 DOI: 10.1371/journal.pone.0075158] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 08/13/2013] [Indexed: 01/06/2023] Open
Abstract
Despite improvements in human leukocyte antigen matching and pharmacologic prophylaxis, acute graft-versus-host disease (GVHD) is often a fatal complication following hematopoietic stem cell transplant (HSCT). Older HSCT recipients experience significantly increased morbidity and mortality compared to young recipients. Prophylaxis with syngeneic regulatory dendritic cells (DCreg) in young bone marrow transplanted (BMT) mice has been shown to decrease GVHD-associated mortality. To evaluate this approach in older BMT recipients, young (3–4 months) and older (14–18 months) DCreg were generated using GM-CSF, IL-10, and TGFβ. Analysis of young versus older DCreg following culture revealed no differences in phenotype. The efficacy of DCreg treatment in older BMT mice was evaluated in a BALB/c→C57Bl/6 model of GVHD; on day 2 post-BMT (d +2), mice received syngeneic, age-matched DCreg. Although older DCreg-treated BMT mice showed decreased morbidity and mortality compared to untreated BMT mice (all of which died), there was a small but significant decrease in the survival of older DCreg-treated BMT mice (75% survival) compared to young DCreg-treated BMT mice (90% survival). To investigate differences between dendritic cells (DC) in young and older DCreg-treated BMT mice that may play a role in DCreg function in vivo, DC phenotypes were assessed following DCreg adoptive transfer. Transferred DCreg identified in older DCreg-treated BMT mice at d +3 showed significantly lower expression of PD-L1 and PIR B compared to DCreg from young DCreg-treated BMT mice. In addition, donor DC identified in d +21 DCreg-treated BMT mice displayed increased inhibitory molecule and decreased co-stimulatory molecule expression compared to d +3, suggesting induction of a regulatory phenotype on the donor DC. In conclusion, these data indicate DCreg treatment is effective in the modulation of GVHD in older BMT recipients and provide evidence for inhibitory pathways that DCreg and donor DC may utilize to induce and maintain tolerance to GVHD.
Collapse
Affiliation(s)
- Sabrina M. Scroggins
- Interdisciplinary Graduate Program in Immunology, University of Iowa Graduate College, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa College of Medicine, Iowa City, Iowa, United States of America
| | - Alicia K. Olivier
- Department of Pathology, University of Iowa College of Medicine, Iowa City, Iowa, United States of America
| | - David K. Meyerholz
- Department of Pathology, University of Iowa College of Medicine, Iowa City, Iowa, United States of America
| | - Annette J. Schlueter
- Interdisciplinary Graduate Program in Immunology, University of Iowa Graduate College, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa College of Medicine, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
80
|
Monsiváis-Urenda A, Gómez-Martin D, Santana-de-Anda K, Cruz-Martínez J, Alcocer-Varela J, González-Amaro R. Defective expression and function of the ILT2/CD85j regulatory receptor in dendritic cells from patients with systemic lupus erythematosus. Hum Immunol 2013; 74:1088-96. [DOI: 10.1016/j.humimm.2013.05.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Revised: 05/06/2013] [Accepted: 05/29/2013] [Indexed: 02/01/2023]
|
81
|
Suppression of SHP-1 promotes corticospinal tract sprouting and functional recovery after brain injury. Cell Death Dis 2013; 4:e567. [PMID: 23559001 PMCID: PMC3641325 DOI: 10.1038/cddis.2013.102] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Reorganization of spared neural network connections is one of the most important processes for restoring impaired function after brain injury. However, plasticity is quite limited in the adult brain due to the presence of inhibitory molecules and a lack of intrinsic neuronal signals for axonal growth. Src homology 2-containing phosphatase (SHP)-1 has been shown to have a role in axon growth inhibition. Here, we tested the hypothesis that SHP-1 negatively affects axonal reorganization. We observed that unilateral motor cortex injury led to increased expression and activity of SHP-1 in the contralesional cortex. In this model, corticospinal axons originating from the contralesional cortex sprouted into the denervated side of the cervical spinal cord after injury. We observed that the number of sprouting fibers was increased in SHP-1-deficient heterozygous viable motheaten (+/mev) mice, which show reduced SHP-1 activity, and in wild-type mice treated with an SHP inhibitor. Motor function recovery of impaired forelimb was enhanced in +/mev mice. Collectively, our results indicate that downregulation of SHP-1 activity promotes corticospinal tract sprouting and functional recovery after brain injury.
Collapse
|
82
|
Yang WC, Ma G, Chen SH, Pan PY. Polarization and reprogramming of myeloid-derived suppressor cells. J Mol Cell Biol 2013; 5:207-9. [PMID: 23532593 DOI: 10.1093/jmcb/mjt009] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) have recently emerged as one of the central regulators of the immune system. In recent years, interest in understanding MDSC biology and applying MDSC for therapeutic purpose has exploded exponentially. Despite recent progress in MDSC biology, the mechanisms underlying MDSC development from expansion and activation to polarization in different diseases remain poorly understood. More recent studies have demonstrated that two MDSC subsets, M (monocytic)-MDSC and G (granulocytic)-MDSC, are able to polarize from a classically activated phenotype (M1) to an alternatively activated one (M2), or vice versa, in tumor-bearing mice. This phenotypic polarization affects MDSC function and disease progression. In this article, we summarize and discuss polarization, mechanism and therapeutic potential of MDSC. An emphasis is placed on the emerging concept of reprogramming MDSC polarization as a therapeutic strategy.
Collapse
Affiliation(s)
- Wen-Chin Yang
- Department of Oncological Sciences and Immunology Institute, Mont Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | |
Collapse
|
83
|
Akkaya M, Barclay AN. How do pathogens drive the evolution of paired receptors? Eur J Immunol 2013; 43:303-13. [DOI: 10.1002/eji.201242896] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 11/06/2012] [Accepted: 12/18/2012] [Indexed: 11/12/2022]
Affiliation(s)
- Munir Akkaya
- Sir William Dunn School of Pathology; University of Oxford; Oxford United Kingdom
| | - A. Neil Barclay
- Sir William Dunn School of Pathology; University of Oxford; Oxford United Kingdom
| |
Collapse
|
84
|
Daws MR, Dai KZ, Zinöcker S, Naper C, Kveberg L, Hedrich HJ, Rolstad B, Vaage JT. Identification of an MHC class I ligand for the single member of a killer cell lectin-like receptor family, KLRH1. THE JOURNAL OF IMMUNOLOGY 2012; 189:5178-84. [PMID: 23100519 DOI: 10.4049/jimmunol.1201983] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Natural killer cells are able to recognize and kill target cells according to differences in MHC class I expression. In rodents, the Ly49 receptors are primarily responsible for this MHC differentiation. We previously described the cloning of a novel C-type lectin-like receptor, KLRH1, encoded in the NK complex adjacent to the Ly49 genes and expressed by subsets of NK and NKT cells. MHC influence on selection of KLRH1(+) NK cells in congenic strains suggested that KLRH1 may have an MHC ligand, although we were unable to identify any such ligand. In this study, we have used a sensitive reporter system and Fc fusion protein to demonstrate that KLRH1 binds specifically to the classical MHC class I molecule RT1-A2 of the RT1(n) haplotype. Cytolytic activity of KLRH1-transfected RNK-16 cells was also inhibited by target cells expressing RT1-A2(n). Thus, KLRH1 represents a novel family of MHC allele-specific inhibitory receptors expressed by NK cells.
Collapse
Affiliation(s)
- Michael R Daws
- Department of Anatomy, University of Oslo, 0317 Oslo, Norway.
| | | | | | | | | | | | | | | |
Collapse
|
85
|
VanGuilder Starkey HD, Van Kirk CA, Bixler GV, Imperio CG, Kale VP, Serfass JM, Farley JA, Yan H, Warrington JP, Han S, Mitschelen M, Sonntag WE, Freeman WM. Neuroglial expression of the MHCI pathway and PirB receptor is upregulated in the hippocampus with advanced aging. J Mol Neurosci 2012; 48:111-26. [PMID: 22562814 DOI: 10.1007/s12031-012-9783-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 04/16/2012] [Indexed: 12/31/2022]
Abstract
The hippocampus undergoes changes with aging that impact neuronal function, such as synapse loss and altered neurotransmitter release. Nearly half of the aged population also develops deficits in spatial learning and memory. To identify age-related hippocampal changes that may contribute to cognitive decline, transcriptomic analysis of synaptosome preparations from adult (12 months) and aged (28 months) Fischer 344-Brown Norway rats assessed for spatial learning and memory was performed. Bioinformatic analysis identified the MHCI pathway as significantly upregulated with aging. Age-related increases in mRNAs encoding the MHCI genes RT1-A1, RT1-A2, and RT1-A3 were confirmed by qPCR in synaptosomes and in CA1 and CA3 dissections. Elevated levels of the MHCI cofactor (B2m), antigen-loading components (Tap1, Tap2, Tapbp), and two known MHCI receptors (PirB, Klra2) were also confirmed. Protein expression of MHCI was elevated with aging in synaptosomes, CA1, and DG, while PirB protein expression was induced in both CA1 and DG. MHCI expression was localized to microglia and neuronal excitatory postsynaptic densities, and PirB was localized to neuronal somata, axons, and dendrites. Induction of the MHCI antigen processing and presentation pathway in hippocampal neurons and glia may contribute to age-related hippocampal dysfunction by increasing neuroimmune signaling or altering synaptic homeostasis.
Collapse
|
86
|
Adelson JD, Barreto GE, Xu L, Kim T, Brott BK, Ouyang YB, Naserke T, Djurisic M, Xiong X, Shatz CJ, Giffard RG. Neuroprotection from stroke in the absence of MHCI or PirB. Neuron 2012; 73:1100-7. [PMID: 22445338 DOI: 10.1016/j.neuron.2012.01.020] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2012] [Indexed: 10/28/2022]
Abstract
Recovery from stroke engages mechanisms of neural plasticity. Here we examine a role for MHC class I (MHCI) H2-Kb and H2-Db, as well as PirB receptor. These molecules restrict synaptic plasticity and motor learning in the healthy brain. Stroke elevates neuronal expression not only of H2-Kb and H2-Db, but also of PirB and downstream signaling. KbDb knockout (KO) or PirB KO mice have smaller infarcts and enhanced motor recovery. KO hippocampal organotypic slices, which lack an intact peripheral immune response, have less cell death after in vitro ischemia. In PirB KO mice, corticospinal projections from the motor cortex are enhanced, and the reactive astrocytic response is dampened after MCAO. Thus, molecules that function in the immune system act not only to limit synaptic plasticity in healthy neurons, but also to exacerbate brain injury after ischemia. These results suggest therapies for stroke by targeting MHCI and PirB.
Collapse
Affiliation(s)
- Jaimie D Adelson
- Department of Biology and Neurobiology, Stanford University, Stanford, CA 94305-5437, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Deng XH, Ai WM, Lei DL, Luo XG, Yan XX, Li Z. Lipopolysaccharide induces paired immunoglobulin-like receptor B (PirB) expression, synaptic alteration, and learning-memory deficit in rats. Neuroscience 2012; 209:161-70. [PMID: 22395112 DOI: 10.1016/j.neuroscience.2012.02.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 01/26/2012] [Accepted: 02/10/2012] [Indexed: 11/19/2022]
Abstract
Some typical immune proteins are expressed in the nervous system, among which the paired-immunoglobulin-like receptor B (PirB) is a receptor for major histocompatibility complex class I antigen (MHC-I), but may play a physiological role in the brain for neuronal circuitry stability by inhibiting synaptic plasticity. Chronic neuroinflammation is common to many neurodegenerative diseases and is often associated with neuronal/synaptic damage and dysfunction. Here we examined the expression of PirB in the rat brain following intracerebral application of lipopolysaccharide (LPS), which has been shown to induce proinflammatory changes and cognitive deficits in rodents. One month after unilateral intrahippocampal LPS injection (10 μg in 4 μl phosphate-buffered saline, PBS), increased protein levels and immunoreactivity of PirB were detected in the ipsilateral hippocampal formation and cortex of the experimental group relative to vehicle (PBS) control. The increased PirB labeling was localized to astrocytes and neurons. Reduced synaptophysin protein levels and immunoreactivity were also found in the ipsilateral hippocampal formation and cortex in LPS-treated rats relative to controls. Morris water maze tests indicated that hippocampus-dependent spatial learning and memory were impaired in LPS-treated animals. Our findings add new experimental data for an upregulation of immune proteins in neuronal and glial cells in the brain in a model of endotoxin-induced neuroinflammation, synaptic alteration, and cognitive decline. The results suggest that PirB modulation may be involved in the pathological process under neurodegenerative conditions.
Collapse
Affiliation(s)
- X-H Deng
- Department of Anatomy and Neurobiology, Central South University, Xiangya School of Medicine, Changsha, Hunan 410013, PR China
| | | | | | | | | | | |
Collapse
|
88
|
Sigalov AB. Interplay Between Protein Order, Disorder and Oligomericity in Receptor Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 725:50-73. [DOI: 10.1007/978-1-4614-0659-4_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
89
|
Challa-Malladi M, Lieu YK, Califano O, Holmes AB, Bhagat G, Murty VV, Dominguez-Sola D, Pasqualucci L, Dalla-Favera R. Combined genetic inactivation of β2-Microglobulin and CD58 reveals frequent escape from immune recognition in diffuse large B cell lymphoma. Cancer Cell 2011; 20:728-40. [PMID: 22137796 PMCID: PMC3660995 DOI: 10.1016/j.ccr.2011.11.006] [Citation(s) in RCA: 350] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 10/21/2011] [Accepted: 11/09/2011] [Indexed: 01/08/2023]
Abstract
We report that diffuse large B cell lymphoma (DLBCL) commonly fails to express cell-surface molecules necessary for the recognition of tumor cells by immune-effector cells. In 29% of cases, mutations and deletions inactivate the β2-Microglobulin gene, thus preventing the cell-surface expression of the HLA class-I (HLA-I) complex that is necessary for recognition by CD8(+) cytotoxic T cells. In 21% of cases, analogous lesions involve the CD58 gene, which encodes a molecule involved in T and natural killer cell-mediated responses. In addition to gene inactivation, alternative mechanisms lead to aberrant expression of HLA-I and CD58 in >60% of DLBCL. These two events are significantly associated in this disease, suggesting that they are coselected during lymphomagenesis for their combined role in escape from immune-surveillance.
Collapse
|
90
|
Hoshino S, Kurishima A, Inaba M, Ando Y, Fukui T, Uchida K, Nishio A, Iwai H, Yokoi T, Ito T, Hasegawa-Ishii S, Shimada A, Li M, Okazaki K, Ikehara S. Amelioration of 2,4,6-trinitrobenzene sulfonic acid-induced colitis in mice by immunoregulatory dendritic cells. J Gastroenterol 2011; 46:1368-81. [PMID: 21922185 DOI: 10.1007/s00535-011-0460-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Accepted: 07/17/2011] [Indexed: 02/04/2023]
Abstract
BACKGROUND Dendritic cells (DCs) are widely distributed throughout the lymphoid and nonlymphoid tissues, and are important initiators of acquired immunity. They also serve as regulators by inducing self-tolerance. However, it has not been thoroughly clarified whether DCs are somehow involved in the regulation or treatment of inflammatory bowel diseases. METHODS We established an ileitis model by transmurally injecting 2,4,6-trinitrobenzene sulfonic acid (TNBS) into the lumen of the ileocolonic junction. The kinetic movement of DCs at the inflammatory sites was analyzed histologically and by flow cytometry, and DCs obtained from the small intestine were analyzed in order to determine the expression of paired immunoglobulin-like receptor-A/B (PIR-A/B) by flow cytometry and quantitative RT-PCR. Furthermore, the regulatory role of DCs was directly determined by a transfer experiment using TNBS-induced colitis model mice. RESULTS We observed three DC subsets (PIR-A/B(high), PIR-A/B(med), and PIR-A/B(-) DCs) in the conventional DCs (cDCs) from day 3, and the number of PIR-A/B(med) cDCs increased from the time the inflammatory responses ceased (day 7). PIR-A/B(med) cDCs actually migrated to the inflamed colon, and ameliorated the colitis induced by TNBS when transferred to colitis-induced recipients. The colitis was greatly exacerbated when mice had been treated with the indoleamine-pyrrole 2,3-dioxygenase (IDO) inhibitor 1-methyltryptophan (1-mT) at the time PIR-A/B(med) cDCs were transferred, indicating that the therapeutic ability of PIR-A/B(med) cDCs is partially dependent on IDO. CONCLUSION The PIR-A/B(med) cDCs, which increase in number during the final stages of inflammation, can be used to treat colitis via an IDO-dependent mechanism.
Collapse
Affiliation(s)
- Shoichi Hoshino
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Moriguchi, Osaka, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Abstract
The Nogo receptor and paired immunoglobulin-like receptor B (PIR-B) are receptors for three myelin-derived axon-growth inhibitors, including myelin-associated glycoprotein (MAG). In this study, we report that the p75 receptor is required for the signal transduction of PIR-B, which interacted with p75 upon ligand binding. In addition, p75 was required for activation of Src homology 2-containing protein tyrosine phosphatase (SHP), which is induced by MAG binding to PIR-B. Mice carrying a mutation in the p75 gene showed promotion of axonal regeneration after optic nerve injury. Thus, our results indicate that p75 has a critical role in axon growth inhibition in specific neuronal tracts.
Collapse
|
92
|
Paired opposing leukocyte receptors recognizing rapidly evolving ligands are subject to homogenization of their ligand binding domains. Immunogenetics 2011; 63:809-20. [PMID: 21720914 PMCID: PMC3210942 DOI: 10.1007/s00251-011-0553-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 06/16/2011] [Indexed: 10/26/2022]
Abstract
Some leukocyte receptors come in groups of two or more where the partners share ligand(s) but transmit opposite signals. Some of the ligands, such as MHC class I, are fast evolving, raising the problem of how paired opposing receptors manage to change in step with respect to ligand binding properties and at the same time conserve opposite signaling functions. An example is the KLRC (NKG2) family, where opposing variants have been conserved in both rodents and primates. Phylogenetic analyses of the KLRC receptors within and between the two orders show that the opposing partners have been subject to post-speciation gene homogenization restricted mainly to the parts of the genes that encode the ligand binding domains. Concerted evolution similarly restricted is demonstrated also for the KLRI, KLRB (NKR-P1), KLRA (Ly49), and PIR receptor families. We propose the term merohomogenization for this phenomenon and discuss its significance for the evolution of immune receptors.
Collapse
|
93
|
Matsushita H, Endo S, Kobayashi E, Sakamoto Y, Kobayashi K, Kitaguchi K, Kuroki K, Söderhäll A, Maenaka K, Nakamura A, Strittmatter SM, Takai T. Differential but competitive binding of Nogo protein and class i major histocompatibility complex (MHCI) to the PIR-B ectodomain provides an inhibition of cells. J Biol Chem 2011; 286:25739-47. [PMID: 21636572 DOI: 10.1074/jbc.m110.157859] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Binding of class I MHC molecules (MHCI) to an inhibitory receptor, PIR-B, expressed on B cells and myeloid cells provides constitutive cellular inhibition, thus ensuring peripheral tolerance. Recent unexpected findings pointed to a novel inhibitory role of PIR-B in neurite regeneration through binding to three axonal outgrowth inhibitors of myelin, including Nogo. Thus, it becomes interesting to determine whether the actions of the inhibitory myelin proteins and MHCI could coexist independently or be mutually exclusive as to the PIR-B-mediated immune and neural cell inhibition. Here, we present data supporting the competition of Nogo- and MHCI-mediated inhibition where they coexist. Kinetic analyses of Nogo and MHCI binding to the whole or a part of the recombinant PIR-B ectodomain revealed that PIR-B binds with higher affinity to Nogo than MHCI and that the MHCI binding only occurred with the N-terminal domains of PIR-B, whereas Nogo binding occurred with either the N- or C-terminal ectodomains. Importantly, kinetic tests indicated that the binding to PIR-B of Nogo and MHCI was competitive. Both endogenous and exogenous Nogo intensified the PIR-B-mediated suppression of interleukin-6 release from lipopolysaccharide-stimulated wild-type, but not PIR-B-deficient, cultured mast cells, indicating that PIR-B mediates Nogo-induced inhibition. Thus, we propose a novel mechanism by which PIR-B-mediated regulation is achieved differentially but competitively via MHCI and Nogo in cells of the immune system.
Collapse
Affiliation(s)
- Haruka Matsushita
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Arita K, Endo S, Kaifu T, Kitaguchi K, Nakamura A, Ohmori H, Kohu K, Satake M, Takai T. Transcriptional Activation of thePirbGene in B Cells by PU.1 and Runx3. THE JOURNAL OF IMMUNOLOGY 2011; 186:7050-9. [DOI: 10.4049/jimmunol.1001302] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
95
|
Lee JK, Zheng B. Role of myelin-associated inhibitors in axonal repair after spinal cord injury. Exp Neurol 2011; 235:33-42. [PMID: 21596039 DOI: 10.1016/j.expneurol.2011.05.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2010] [Revised: 04/19/2011] [Accepted: 05/01/2011] [Indexed: 12/15/2022]
Abstract
Myelin-associated inhibitors of axon growth, including Nogo, MAG and OMgp, have been the subject of intense research. A myriad of experimental approaches have been applied to investigate the potential of targeting these molecules to promote axonal repair after spinal cord injury. However, there are still conflicting results on their role in axon regeneration and therefore a lack of a cohesive mechanism on how these molecules can be targeted to promote axon repair. One major reason may be the lack of a clear definition of axon regeneration in the first place. Nevertheless, recent data from genetic studies in mice indicate that the roles of these molecules in CNS axon repair may be more intricate than previously envisioned.
Collapse
Affiliation(s)
- Jae K Lee
- University of California San Diego, Department of Neurosciences, 9500 Gilman Drive, MC 0691, La Jolla, CA 92093-0691, USA.
| | | |
Collapse
|
96
|
Ma G, Pan PY, Eisenstein S, Divino CM, Lowell CA, Takai T, Chen SH. Paired immunoglobin-like receptor-B regulates the suppressive function and fate of myeloid-derived suppressor cells. Immunity 2011; 34:385-95. [PMID: 21376641 DOI: 10.1016/j.immuni.2011.02.004] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 11/23/2010] [Accepted: 02/01/2011] [Indexed: 12/12/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) bear characteristics of precursors for both M1 and M2 macrophages. The molecular mechanism underlying the differentiation into M1 and M2 macrophages and the relationship of this differentiation to antitumor responses remains largely undefined. Herein, we investigate the potential function of paired immunoglobulin-like receptor B (PIR-B), also known as leukocyte immunoglobulin-like receptor subfamily B member 3 (LILRB3) in MDSC differentiation, and its role in tumor-induced immunity. Our studies indicated that MDSCs genetically ablated for PIR-B (Lilrb3(-/-)) underwent a specific transition to M1-like cells when entering the periphery from bone marrow, resulting in decreased suppressive function, regulatory T cell activation activity, primary tumor growth, and lung metastases. Activation of Toll-like receptor (TLR), signal transducers, and activators of transcription 1 (STAT1), and nuclear factor-kappa B (NF-κB) signaling in Lilrb3(-/-) MDSC promoted the acquisition of M1 phenotype. Inhibition of the PIR-B signaling pathway promoted MDSC differentiation into M1 macrophages.
Collapse
Affiliation(s)
- Ge Ma
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, 1425 Madison Avenue, Room 13-02, New York, NY 10029-6574, USA
| | | | | | | | | | | | | |
Collapse
|
97
|
Fujita Y, Endo S, Takai T, Yamashita T. Myelin suppresses axon regeneration by PIR-B/SHP-mediated inhibition of Trk activity. EMBO J 2011; 30:1389-401. [PMID: 21364532 DOI: 10.1038/emboj.2011.55] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 02/04/2011] [Indexed: 11/10/2022] Open
Abstract
Paired immunoglobulin-like receptor B (PIR-B) partially mediates the regeneration-inhibiting effects of the myelin-derived protein Nogo, myelin-associated glycoprotein (MAG), and oligodendrocyte-myelin glycoprotein (OMgp). In this study, we report that inhibition of the PIR-B signaling cascades in neurons enhances axon regeneration in the central nervous system (CNS). Binding of MAG to PIR-B led to the association of PIR-B with tropomyosin receptor kinase (Trk) neurotrophin receptors. Src homology 2-containing protein tyrosine phosphatase (SHP)-1 and SHP-2, which were recruited to PIR-B upon MAG binding, functioned as Trk tyrosine phosphatases. Further, SHP-1 and SHP-2 inhibition reduced MAG-induced dephosphorylation of Trk receptors and abolished the inhibitory effect of MAG on neurite growth. Thus, PIR-B associated with Trk to downregulate basal and neurotrophin-regulated Trk activity through SHP-1/2 in neurons. Moreover, in vivo transfection of small interfering RNA (siRNA) for SHP-1 or SHP-2 induced axonal regeneration after optic nerve injury in mice. Our results thus identify a new molecular target to enhance regeneration of the injured CNS.
Collapse
Affiliation(s)
- Yuki Fujita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan
| | | | | | | |
Collapse
|
98
|
Steevels TAM, Meyaard L. Immune inhibitory receptors: essential regulators of phagocyte function. Eur J Immunol 2011; 41:575-87. [PMID: 21312193 DOI: 10.1002/eji.201041179] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 01/06/2011] [Accepted: 01/20/2011] [Indexed: 01/21/2023]
Abstract
Phagocytes, including neutrophils, monocytes, and macrophages, play a crucial role in host defense by recognition and elimination of invading pathogens. Phagocytic cells produce reactive oxygen species (ROS), inflammatory cytokines, and chemokines, leading to bacterial killing and to recruitment and activation of additional immune cells. However, inflammatory mediators are potentially harmful for the host and their production is therefore tightly controlled by multiple regulatory mechanisms. One such mechanism is immune suppression by immune inhibitory receptors, which are increasingly acknowledged as potent regulators of the immune response. So far, research has focused on the role of these receptors in the regulation of NK cells, B cells, and T cells. Importantly, an accumulating number of inhibitory receptors have been identified on phagocytes. Here, we review the role of inhibitory receptors in the regulation of phagocyte cytokine production, migration, apoptosis, ROS production, and phagocytosis. Furthermore, we discuss the intracellular mechanisms utilized by distinct inhibitory receptors to regulate specific phagocyte functions. We demonstrate that inhibitory receptors are important regulators of the immune response, which bacteria can use to their advantage.
Collapse
Affiliation(s)
- Tessa A M Steevels
- Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
99
|
Fourgeaud L, Boulanger LM. Role of immune molecules in the establishment and plasticity of glutamatergic synapses. Eur J Neurosci 2011; 32:207-17. [PMID: 20946111 DOI: 10.1111/j.1460-9568.2010.07342.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An increasing number of studies support an unexpected role for immune molecules in regulating healthy brain functions during development and in adulthood. Here we review the roles of specific immune molecules (including cytokines, components of the complement cascade, and members of the major histocompatibility complex class I family and their receptors) in the formation and plasticity of glutamatergic synapses. These findings add a new dimension to our understanding of neural-immune interactions, and suggest novel molecular mechanisms that may underlie the modification of glutamatergic synapses in both normal and pathological states.
Collapse
Affiliation(s)
- Lawrence Fourgeaud
- Molecular Neurobiology Laboratories (MNL-L), The Salk Institute for Biological Studies, La Jolla, CA, USA
| | | |
Collapse
|
100
|
The phylogenetic origins of natural killer receptors and recognition: relationships, possibilities, and realities. Immunogenetics 2010; 63:123-41. [PMID: 21191578 DOI: 10.1007/s00251-010-0506-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 12/16/2010] [Indexed: 12/20/2022]
Abstract
Natural killer (NK) cells affect a form of innate immunity that recognizes and eliminates cells that are infected with certain viruses or have undergone malignant transformation. In mammals, this recognition can be mediated through immunoglobulin- (Ig) and/or lectin-type NK receptors (NKRs). NKR genes in mammals range from minimally polymorphic single-copy genes to complex multigene families that exhibit high levels of haplotypic complexity and exhibit significant interspecific variation. Certain single-copy NKR genes that are present in one mammal are present as expanded multigene families in other mammals. These observations highlight NKRs as one of the most rapidly evolving eukaryotic gene families and likely reflect the influence of pathogens, especially viruses, on their evolution. Although well characterized in human and mice, cytotoxic cells that are functionally similar to NK cells have been identified in species ranging from birds to reptiles, amphibians and fish. Although numerous receptors have been identified in non-mammalian vertebrates that share structural relationships with mammalian NKRs, functionally defining these lower vertebrate molecules as NKRs is confounded by methodological and interpretive complexities. Nevertheless, several lines of evidence suggest that NK-type function or its equivalent has sustained a long evolutionary history throughout vertebrate species.
Collapse
|