51
|
Patel M, Vlahava VM, Forbes SK, Fielding CA, Stanton RJ, Wang ECY. HCMV-Encoded NK Modulators: Lessons From in vitro and in vivo Genetic Variation. Front Immunol 2018; 9:2214. [PMID: 30327650 PMCID: PMC6174198 DOI: 10.3389/fimmu.2018.02214] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 09/06/2018] [Indexed: 12/22/2022] Open
Abstract
Human cytomegalovirus (HCMV) is under constant selective pressure from the immune system in vivo. Study of HCMV genes that have been lost in the absence of, or genetically altered by, such selection can focus research toward findings of in vivo significance. We have been particularly interested in the most pronounced change in the highly passaged laboratory strains AD169 and Towne—the deletion of 13–15 kb of sequence (designated the UL/b′ region) that encodes up to 22 canonical genes, UL133-UL150. At least 5 genes have been identified in UL/b′ that inhibit NK cell function. UL135 suppresses formation of the immunological synapse (IS) by remodeling the actin cytoskeleton, thereby illustrating target cell cooperation in IS formation. UL141 inhibits expression of two activating ligands (CD155, CD112) for the activating receptor CD226 (DNAM-1), and two receptors (TRAIL-R1, R2) for the apoptosis-inducing TRAIL. UL142, ectopically expressed in isolation, and UL148A, target specific MICA allotypes that are ligands for NKG2D. UL148 impairs expression of CD58 (LFA-3), the co-stimulatory cell adhesion molecule for CD2 found on T and NK cells. Outside UL/b′, studies on natural variants have shown UL18 mutants change affinity for their inhibitory ligand LIR-1, while mutations in UL40's HLA-E binding peptide differentially drive NKG2C+ NK expansions. Research into HCMV genomic stability and its effect on NK function has provided important insights into virus:host interactions, but future studies will require consideration of genetic variability and the effect of genes expressed in the context of infection to fully understand their in vivo impact.
Collapse
Affiliation(s)
- Mihil Patel
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Virginia-Maria Vlahava
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Simone K Forbes
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Ceri A Fielding
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Richard J Stanton
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Eddie C Y Wang
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| |
Collapse
|
52
|
Sun H, Xu J, Huang Q, Huang M, Li K, Qu K, Wen H, Lin R, Zheng M, Wei H, Xiao W, Sun R, Tian Z, Sun C. Reduced CD160 Expression Contributes to Impaired NK-cell Function and Poor Clinical Outcomes in Patients with HCC. Cancer Res 2018; 78:6581-6593. [PMID: 30232222 DOI: 10.1158/0008-5472.can-18-1049] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 08/09/2018] [Accepted: 09/10/2018] [Indexed: 11/16/2022]
Abstract
: We previously reported that deficiencies in natural killer (NK)-cell number and function play an important role in the progression of hepatocellular carcinoma (HCC). However, the mechanisms underlying this phenomenon remain obscure. In this study, we analyzed the expression of CD160 on intrahepatic NK cells by evaluating peritumoral and intratumoral tissues of 279 patients with HCC and 20 healthy livers. We observed reduced expression of CD160 on intratumoral NK cells, and patients with lower CD160 cell densities within tumors exhibited worse disease and a higher recurrence rate. High-resolution microarray and gene set enrichment analysis of flow cytometry-sorted primary intrahepatic CD160+ and CD160- NK cells of healthy livers indicated that human CD160+ NK cells exhibited functional activation, high IFNγ production, and NK-mediated immunity. In addition, global transcriptomic analysis of sorted peritumoral and intratumoral CD160+ NK cells revealed that intratumoral CD160+ NK cells are more exhausted than peritumoral CD160+ NK cells and produce less IFNγ. High levels of TGFβ1 interfered with production of IFNγ by CD160+ NK cells, blocking of which specifically restored IFNγ production in CD160+ NK cells to normal levels. These findings indicate that reduced numbers of CD160+ NK cells, together with the functional impairment of CD160+ NK cells by TGFβ1, contribute to tumor immune escape. In addition, restoring the expression of CD160 and blocking TGFβ1 appear a promising therapeutic strategy against liver cancer. SIGNIFICANCE: These findings show that reduced number and function of CD160+ NK cells in the tumor microenvironment contributes to immune escape of HCC; blocking TGFβ1 restores IFNγ production of CD160+ NK cells.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/23/6581/F1.large.jpg.
Collapse
Affiliation(s)
- Haoyu Sun
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Jing Xu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Qiang Huang
- Organ Transplant Center & Immunology Laboratory, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Mei Huang
- Organ Transplant Center & Immunology Laboratory, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Kun Li
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Kun Qu
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Hao Wen
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Renyong Lin
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Meijuan Zheng
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Haiming Wei
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Weihua Xiao
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Rui Sun
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China.,Xinjiang Key Laboratory of Echinococcosis, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Zhigang Tian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China. .,Institute of Immunology, University of Science and Technology of China, Hefei, China.,Xinjiang Key Laboratory of Echinococcosis, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Cheng Sun
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China. .,Institute of Immunology, University of Science and Technology of China, Hefei, China.,Xinjiang Key Laboratory of Echinococcosis, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
53
|
Šedý JR, Balmert MO, Ware BC, Smith W, Nemčovičova I, Norris PS, Miller BR, Aivazian D, Ware CF. A herpesvirus entry mediator mutein with selective agonist action for the inhibitory receptor B and T lymphocyte attenuator. J Biol Chem 2017; 292:21060-21070. [PMID: 29061848 DOI: 10.1074/jbc.m117.813295] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/04/2017] [Indexed: 12/19/2022] Open
Abstract
The human cytomegalovirus opening reading frame UL144 is an ortholog of the TNF receptor superfamily member, herpesvirus entry mediator (HVEM; TNFRSF14). HVEM binds the TNF ligands, LIGHT and LTa; the immunoglobulin inhibitory receptor, B and T lymphocyte attenuator (BTLA); and the natural killer cell-activating receptor CD160. However, UL144 selectively binds BTLA, avoiding activation of inflammatory signaling initiated by CD160 in natural killer cells. BTLA and CD160 cross-compete for binding HVEM, but the structural basis for the ligand selectivity by UL144 and how it acts as an anti-inflammatory agonist remains unclear. Here, we modeled the UL144 structure and characterized its binding with BTLA. The UL144 structure was predicted to closely mimic the surface of HVEM, and we also found that both HVEM and UL144 bind a common epitope of BTLA, whether engaged in trans or in cis, that is shared with a BTLA antibody agonist. On the basis of the UL144 selectivity, we engineered a BTLA-selective HVEM protein to understand the basis for ligand selectivity and BTLA agonism to develop novel anti-inflammatory agonists. This HVEM mutein did not bind CD160 or TNF ligands but did bind BTLA with 10-fold stronger affinity than wild-type HVEM and retained potent inhibitory activity that reduced T-cell receptor, B-cell receptor, and interferon signaling in B cells. In conclusion, using a viral immune evasion strategy that shows broad immune-ablating activity, we have identified a novel anti-inflammatory BTLA-selective agonist.
Collapse
Affiliation(s)
- John R Šedý
- From the Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037,
| | - M Olivia Balmert
- From the Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037
| | - Brian C Ware
- From the Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037
| | - Wendell Smith
- From the Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037
| | - Ivana Nemčovičova
- the Biomedical Research Center, Slovak Academy of Sciences, SK 84505, Bratislava, Slovakia, and
| | - Paula S Norris
- From the Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037
| | - Brian R Miller
- the Centers for Therapeutic Innovation, Pfizer Inc., La Jolla, California 92037
| | - Dikran Aivazian
- the Centers for Therapeutic Innovation, Pfizer Inc., La Jolla, California 92037
| | - Carl F Ware
- From the Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037,
| |
Collapse
|
54
|
Ward-Kavanagh LK, Lin WW, Šedý JR, Ware CF. The TNF Receptor Superfamily in Co-stimulating and Co-inhibitory Responses. Immunity 2017; 44:1005-19. [PMID: 27192566 DOI: 10.1016/j.immuni.2016.04.019] [Citation(s) in RCA: 301] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Indexed: 02/08/2023]
Abstract
Cytokines related to tumor necrosis factor (TNF) provide a communication network essential for coordinating multiple cell types into an effective host defense system against pathogens and malignant cells. The pathways controlled by the TNF superfamily differentiate both innate and adaptive immune cells and modulate stromal cells into microenvironments conducive to host defenses. Members of the TNF receptor superfamily activate diverse cellular functions from the production of type 1 interferons to the modulation of survival of antigen-activated T cells. Here, we focus attention on the subset of TNF superfamily receptors encoded in the immune response locus in chromosomal region 1p36. Recent studies have revealed that these receptors use diverse mechanisms to either co-stimulate or restrict immune responses. Translation of the fundamental mechanisms of TNF superfamily is leading to the design of therapeutics that can alter pathogenic processes in several autoimmune diseases or promote immunity to tumors.
Collapse
Affiliation(s)
- Lindsay K Ward-Kavanagh
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Wai Wai Lin
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - John R Šedý
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Carl F Ware
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
55
|
Edwards RG, Longnecker R. Herpesvirus Entry Mediator and Ocular Herpesvirus Infection: More than Meets the Eye. J Virol 2017; 91:e00115-17. [PMID: 28404853 PMCID: PMC5469272 DOI: 10.1128/jvi.00115-17] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
As its name suggests, the host receptor herpesvirus entry mediator (HVEM) facilitates herpes simplex virus (HSV) entry through interactions with a viral envelope glycoprotein. HVEM also bridges several signaling networks, binding ligands from both tumor necrosis factor (TNF) and immunoglobulin (Ig) superfamilies with diverse, and often opposing, outcomes. While HVEM was first identified as a viral entry receptor for HSV, it is only recently that HVEM has emerged as an important host factor in immunopathogenesis of ocular HSV type 1 (HSV-1) infection. Surprisingly, HVEM exacerbates disease development in the eye independently of entry. HVEM signaling has been shown to play a variety of roles in modulating immune responses to HSV and other pathogens, and there is increasing evidence that these effects are responsible for HVEM-mediated pathogenesis in the eye. Here, we review the dual branches of HVEM function during HSV infection: entry and immunomodulation. HVEM is broadly expressed; intersects two important immunologic signaling networks; and impacts autoimmunity, infection, and inflammation. We hope that by understanding the complex range of effects mediated by this receptor, we can offer insights applicable to a wide variety of disease states.
Collapse
Affiliation(s)
- Rebecca G Edwards
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Richard Longnecker
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
56
|
Del Rio ML, Bravo Moral AM, Fernandez-Renedo C, Buhler L, Perez-Simon JA, Chaloin O, Alvarez Nogal R, Fernandez-Caso M, Rodriguez-Barbosa JI. Modulation of cytotoxic responses by targeting CD160 prolongs skin graft survival across major histocompatibility class I barrier. Transl Res 2017; 181:83-95.e3. [PMID: 27702550 DOI: 10.1016/j.trsl.2016.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 08/23/2016] [Accepted: 09/08/2016] [Indexed: 11/26/2022]
Abstract
CD160 is a glycosylphosphatidylinositol-anchored protein of the immunoglobulin superfamily. It exhibits a pattern of expression coincident in humans and mice that is mainly restricted to cytotoxic cells and to all intestinal intraepithelial T lymphocytes. B- and T-lymphocyte attenuator (BTLA) and CD160 interact with cysteine-rich domain 1 of the extracellular region of Herpesvirus entry mediator (HVEM). CD160 engagement by HVEM can deliver inhibitory signals to a small subset of human CD4 T cells and attenuate its proliferation and cytokine secretion, but can also costimulate natural killer cells or intraepithelial lymphocytes. In turn, CD160 and BTLA can also function as agonist ligands being capable of costimulating T cells through membrane HVEM. Based on the restricted pattern of CD160 expression in cytotoxic cells, we postulated that CD160 may represent a suitable target for immune intervention in the setting of transplantation to modulate allogeneic cytotoxic responses. We demonstrated that in vivo administration of anti-CD160 antibody in combination with anti-CD40 L antibody to limit CD4 T-cell help modulated cytotoxic responses in a major histocompatibility complex class I mismatched model of allogeneic skin graft transplantation (bm1 donor to C57BL/6 recipient) and significantly prolonged graft survival. The implementation of this strategy in transplantation may reinforce current immunosuppression protocols and contribute to a better control of CD8 T-cell responses.
Collapse
Affiliation(s)
- Maria-Luisa Del Rio
- Transplantation Immunobiology Section, University of Leon and Castilla and Leon Regional Transplantation Coordination, Leon University Hospital, Leon, Spain.
| | - Ana Maria Bravo Moral
- Department of Veterinary Clinical Sciences, University of Santiago de Compostela, Veterinary Faculty, Lugo, Spain
| | - Carlos Fernandez-Renedo
- Transplantation Immunobiology Section, University of Leon and Castilla and Leon Regional Transplantation Coordination, Leon University Hospital, Leon, Spain
| | - Leo Buhler
- Visceral and Transplantation Surgery, Department of Surgery, University Hospitals of Geneva and Faculty of Medicine, Geneva, Switzerland
| | - Jose-Antonio Perez-Simon
- Department of Hematology, University Hospital Virgen del Rocio/Institute of Biomedicine (IBIS/CSIC), Sevilla, Spain
| | - Olivier Chaloin
- CNRS UPR 3572, IBMC, Immunopathologie et Chimie Thérapeutique, Strasbourg, France
| | - Rafael Alvarez Nogal
- Department of Molecular and Cell Biology, School of Biological Sciences, University of Leon, Leon, Spain
| | - Maximino Fernandez-Caso
- Department of Medicine, Surgery and Veterinary Anatomy, School of Veterinary Medicine, University of Leon, Leon, Spain
| | - Jose-Ignacio Rodriguez-Barbosa
- Transplantation Immunobiology Section, University of Leon and Castilla and Leon Regional Transplantation Coordination, Leon University Hospital, Leon, Spain.
| |
Collapse
|
57
|
Vaccari M, Gordon SN, Fourati S, Schifanella L, Liyanage NPM, Cameron M, Keele BF, Shen X, Tomaras GD, Billings E, Rao M, Chung AW, Dowell KG, Bailey-Kellogg C, Brown EP, Ackerman ME, Vargas-Inchaustegui DA, Whitney S, Doster MN, Binello N, Pegu P, Montefiori DC, Foulds K, Quinn DS, Donaldson M, Liang F, Loré K, Roederer M, Koup RA, McDermott A, Ma ZM, Miller CJ, Phan TB, Forthal DN, Blackburn M, Caccuri F, Bissa M, Ferrari G, Kalyanaraman V, Ferrari MG, Thompson D, Robert-Guroff M, Ratto-Kim S, Kim JH, Michael NL, Phogat S, Barnett SW, Tartaglia J, Venzon D, Stablein DM, Alter G, Sekaly RP, Franchini G. Adjuvant-dependent innate and adaptive immune signatures of risk of SIVmac251 acquisition. Nat Med 2016; 22:762-70. [PMID: 27239761 PMCID: PMC5916782 DOI: 10.1038/nm.4105] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 04/05/2016] [Indexed: 12/24/2022]
Abstract
A recombinant vaccine containing Aventis Pasteur's canarypox vector (ALVAC)-HIV and gp120 alum decreased the risk of HIV acquisition in the RV144 vaccine trial. The substitution of alum with the more immunogenic MF59 adjuvant is under consideration for the next efficacy human trial. We found here that an ALVAC-simian immunodeficiency virus (SIV) and gp120 alum (ALVAC-SIV + gp120) equivalent vaccine, but not an ALVAC-SIV + gp120 MF59 vaccine, was efficacious in delaying the onset of SIVmac251 in rhesus macaques, despite the higher immunogenicity of the latter adjuvant. Vaccine efficacy was associated with alum-induced, but not with MF59-induced, envelope (Env)-dependent mucosal innate lymphoid cells (ILCs) that produce interleukin (IL)-17, as well as with mucosal IgG to the gp120 variable region 2 (V2) and the expression of 12 genes, ten of which are part of the RAS pathway. The association between RAS activation and vaccine efficacy was also observed in an independent efficacious SIV-vaccine approach. Whether RAS activation, mucosal ILCs and antibodies to V2 are also important hallmarks of HIV-vaccine efficacy in humans will require further studies.
Collapse
Affiliation(s)
- Monica Vaccari
- Animal Models and Vaccine Section, National Cancer Institute, Bethesda, Maryland, USA
| | - Shari N Gordon
- Animal Models and Vaccine Section, National Cancer Institute, Bethesda, Maryland, USA
| | - Slim Fourati
- Department of Pathology, Case Western Reserve, Cleveland, Ohio, USA
| | - Luca Schifanella
- Animal Models and Vaccine Section, National Cancer Institute, Bethesda, Maryland, USA
- Department of Biomedical and Clinical Sciences, 'L. Sacco' Hospital, University of Milan, Italy
| | - Namal P M Liyanage
- Animal Models and Vaccine Section, National Cancer Institute, Bethesda, Maryland, USA
| | - Mark Cameron
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland, USA
| | - Xiaoying Shen
- Duke Human Vaccine Institute, Durham, North Carolina, USA
| | | | - Erik Billings
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Mangala Rao
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Amy W Chung
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Boston, Massachusetts, USA
| | - Karen G Dowell
- Department of Computer Science, Dartmouth College, Hanover, New Hampshire, USA
| | | | - Eric P Brown
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Margaret E Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | | | | | - Melvin N Doster
- Animal Models and Vaccine Section, National Cancer Institute, Bethesda, Maryland, USA
| | - Nicolo Binello
- Animal Models and Vaccine Section, National Cancer Institute, Bethesda, Maryland, USA
| | - Poonam Pegu
- Animal Models and Vaccine Section, National Cancer Institute, Bethesda, Maryland, USA
| | | | - Kathryn Foulds
- Vaccine Research Center, US National Institutes of Health, Bethesda, Maryland, USA
| | - David S Quinn
- Vaccine Research Center, US National Institutes of Health, Bethesda, Maryland, USA
| | - Mitzi Donaldson
- Vaccine Research Center, US National Institutes of Health, Bethesda, Maryland, USA
| | | | | | - Mario Roederer
- Vaccine Research Center, US National Institutes of Health, Bethesda, Maryland, USA
| | - Richard A Koup
- Vaccine Research Center, US National Institutes of Health, Bethesda, Maryland, USA
| | - Adrian McDermott
- Vaccine Research Center, US National Institutes of Health, Bethesda, Maryland, USA
| | - Zhong-Min Ma
- California National Primate Research Center, University of California, Davis, California, USA
| | - Christopher J Miller
- California National Primate Research Center, University of California, Davis, California, USA
| | - Tran B Phan
- University of California, Irvine School of Medicine, Irvine, California, USA
| | - Donald N Forthal
- University of California, Irvine School of Medicine, Irvine, California, USA
| | - Matthew Blackburn
- Animal Models and Vaccine Section, National Cancer Institute, Bethesda, Maryland, USA
| | - Francesca Caccuri
- Animal Models and Vaccine Section, National Cancer Institute, Bethesda, Maryland, USA
| | - Massimiliano Bissa
- Animal Models and Vaccine Section, National Cancer Institute, Bethesda, Maryland, USA
| | - Guido Ferrari
- Duke Human Vaccine Institute, Durham, North Carolina, USA
| | | | | | - DeVon Thompson
- Advanced Bioscience Laboratories, Rockville, Maryland, USA
| | - Marjorie Robert-Guroff
- Immune Biology of Retroviral Infection Section, National Cancer Institute, Bethesda, Maryland, USA
| | - Silvia Ratto-Kim
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Jerome H Kim
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Nelson L Michael
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | | | | | | | - David Venzon
- Biostatistics and Data Management Section, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Galit Alter
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Boston, Massachusetts, USA
| | | | - Genoveffa Franchini
- Animal Models and Vaccine Section, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
58
|
Shemesh A, Brusilovsky M, Hadad U, Teltsh O, Edri A, Rubin E, Campbell KS, Rosental B, Porgador A. Survival in acute myeloid leukemia is associated with NKp44 splice variants. Oncotarget 2016; 7:32933-45. [PMID: 27102296 PMCID: PMC5078064 DOI: 10.18632/oncotarget.8782] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 03/27/2016] [Indexed: 11/25/2022] Open
Abstract
NKp44 is a receptor encoded by the NCR2 gene, which is expressed by cytokine-activated natural killer (NK) cells that are involved in anti-AML immunity. NKp44 has three splice variants corresponding to NKp44ITIM+ (NKp44-1) and NKp44ITIM- (NKp44-2, and NKp44-3) isoforms. RNAseq data of AML patients revealed similar survival of NKp46+NKp44+ and NKp46+NKp44- patients. However, if grouped according to the NKp44 splice variant profile, NKp44-1 expression was significantly associated with poor survival of AML patients. Moreover, activation of PBMC from healthy controls showed co-dominant expression of NKp44-1 and NKp44-3, while primary NK clones show more diverse NKp44 splice variant profiles. Cultured primary NK cells resulted in NKp44-1 dominance and impaired function associated with PCNA over-expression by target cells. This impaired functional phenotype could be rescued by blocking of NKp44 receptor. Human NK cell lines revealed co-dominant expression of NKp44-1 and NKp44-3 and showed a functional phenotype that was not inhibited by PCNA over-expression. Furthermore, transfection-based overexpression of NKp44-1, but not NKp44-2/NKp44-3, reversed the endogenous resistance of NK-92 cells to PCNA-mediated inhibition, and resulted in poor formation of stable lytic immune synapses. This research contributes to the understanding of AML prognosis by shedding new light on the functional implications of differential splicing of NKp44.
Collapse
Affiliation(s)
- Avishai Shemesh
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Michael Brusilovsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Uzi Hadad
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Omri Teltsh
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Avishay Edri
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Eitan Rubin
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Kerry S. Campbell
- Immune Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Benyamin Rosental
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine and the Hopkins Marine Station, Stanford, CA, USA
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
59
|
Herpesvirus entry mediator on radiation-resistant cell lineages promotes ocular herpes simplex virus 1 pathogenesis in an entry-independent manner. mBio 2015; 6:e01532-15. [PMID: 26489863 PMCID: PMC4620471 DOI: 10.1128/mbio.01532-15] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Ocular herpes simplex virus 1 (HSV-1) infection leads to a potentially blinding immunoinflammatory syndrome, herpes stromal keratitis (HSK). Herpesvirus entry mediator (HVEM), a widely expressed tumor necrosis factor (TNF) receptor superfamily member with diverse roles in immune signaling, facilitates viral entry through interactions with viral glycoprotein D (gD) and is important for HSV-1 pathogenesis. We subjected mice to corneal infection with an HSV-1 mutant in which HVEM-mediated entry was specifically abolished and found that the HVEM-entry mutant produced clinical disease comparable to that produced by the control virus. HVEM-mediated induction of corneal cytokines, which correlated with an HVEM-dependent increase in levels of corneal immune cell infiltrates, was also gD independent. Given the complexity of HVEM immune signaling, we used hematopoietic chimeric mice to determine which HVEM-expressing cells mediate HSV-1 pathogenesis in the eye. Regardless of whether the donor was a wild-type (WT) or HVEM knockout (KO) strain, HVEM KO recipients were protected from ocular HSV-1, suggesting that HVEM on radiation-resistant cell types, likely resident cells of the cornea, confers wild-type-like susceptibility to disease. Together, these data indicate that HVEM contributes to ocular pathogenesis independently of entry and point to an immunomodulatory role for this protein specifically on radiation-resistant cells. Immune privilege is maintained in the eye in order to protect specialized ocular tissues, such as the translucent cornea, from vision-reducing damage. Ocular herpes simplex virus 1 (HSV-1) infection can disrupt this immune privilege, provoking a host response that ultimately brings about the majority of the damage seen with the immunoinflammatory syndrome herpes stromal keratitis (HSK). Our previous work has shown that HVEM, a host TNF receptor superfamily member that also serves as a viral entry receptor, is a critical component contributing to ocular HSV-1 pathogenesis, although its precise role in this process remains unclear. We hypothesized that HVEM promotes an inflammatory microenvironment in the eye through immunomodulatory actions, enhancing disease after ocular inoculation of HSV-1. Investigating the mechanisms responsible for orchestrating this aberrant immune response shed light on the initiation and maintenance of HSK, one of the leading causes of infectious blindness in the developed world.
Collapse
|
60
|
Lv M, Wu W, Zhang Y, Zhu M. Herpes virus entry mediator licenses Listeria infection induced immunopathology through control of type I interferon. Sci Rep 2015; 5:12954. [PMID: 26245828 PMCID: PMC4526852 DOI: 10.1038/srep12954] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/15/2015] [Indexed: 01/28/2023] Open
Abstract
Apoptosis of the splenic lymphocytes is often induced during the acute phase of Listeria infection in mice. However, the underlying mechanism remains incompletely understood. Here, we found that herpes virus entry mediator (HVEM) plays an important role for Listeria infection induced lymphocyte apoptosis. Mechanistically, HVEM is not directly involved in listeriolysin O (LLO) induced lymphocyte apoptosis or interferon beta induced T cell activation per se. Interestingly, HVEM is partially required for Listeria induced interferon (IFN)-I production in the spleen, particularly in macrophages. Consequently, the bystander activation of lymphocytes is significantly lower in HVEM deficient mice than that in wild-type (WT) mice upon Listeria infection. Thus, our results have revealed a novel role of HVEM on the regulation of IFN-I and immunopathology during Listeria infection.
Collapse
Affiliation(s)
- Mengjie Lv
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Weiwei Wu
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuejiao Zhang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Mingzhao Zhu
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
61
|
Liu W, Vigdorovich V, Zhan C, Patskovsky Y, Bonanno JB, Nathenson SG, Almo SC. Increased Heterologous Protein Expression in Drosophila S2 Cells for Massive Production of Immune Ligands/Receptors and Structural Analysis of Human HVEM. Mol Biotechnol 2015. [DOI: 10.1007/s12033-015-9881-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
62
|
Zuo J, Shan Z, Zhou L, Yu J, Liu X, Gao Y. Increased CD160 expression on circulating natural killer cells in atherogenesis. J Transl Med 2015; 13:188. [PMID: 26071079 PMCID: PMC4467674 DOI: 10.1186/s12967-015-0564-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/03/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Atherosclerosis (AS) presents characteristic of a chronic inflammatory disease in which both adaptive and innate immune cells play roles. Accumulating evidence has showed the impairment of natural killer (NK) cells in atherosclerosis, however, the mechanisms of this impairment remain unclear. In this study, we investigated the expression of CD160 on NK cells and assessed its pathological roles in NK loss during atherogenesis. METHODS CD160 expression on NK cells was measured in 49 AS patients and 41 healthy controls (HC) by flow cytometry, their inflammatory cytokine levels in sera were determined by ELSIA, and the effect of CD160 engagement on NK cells was evaluated by in vitro culture experiments. RESULTS Compared to HC, AS patients had a significantly increased CD160 expression on peripheral NK cells and concomitantly decreased peripheral NK cell number, and increased CD160 expression was positively related to the levels of serum lipids and IFN-γ, TNF-α and IL-6 inflammation cytokines, which all are risk factors for atherogenesis, and inversely correlated with peripheral NK cell number. Furthermore, engagement of CD160 receptor on NK cells from AS patients triggers a significantly increased production of inflammation cytokines and subsequent NK cell apoptosis, and blockade of TNF-α prevented the increased apoptosis of NK cells from AS patients after CD160 engagement, indicating a critical role of TNF-α in mediating NK cell loss by CD160 engagement. RESULTS Our results provide evidence that elevated CD160 expression on NK cells plays an important role in NK cell loss in atherosclerosis. The increased CD160 expression on NK cells might be used as an indicator for disease progression.
Collapse
Affiliation(s)
- Jin Zuo
- Department of Cardiology, Navy General Hospital of Chinese PLA, Beijing, 100863, China.
| | - Zhaoliang Shan
- Department of Cardiology, General Hospital of Chinese PLA, Beijing, 100853, China.
| | - Lin Zhou
- Department of Interventional Radiology, 302 Hospital of Chinese PLA, Beijing, 100039, China.
| | - Jian Yu
- Center of Health Examination, Navy General Hospital of Chinese PLA, Beijing, 100048, China.
| | - Xiaopeng Liu
- Center of Health Examination, Navy General Hospital of Chinese PLA, Beijing, 100048, China.
| | - Yuan Gao
- Department of Cardiology, Navy General Hospital of Chinese PLA, Beijing, 100863, China.
| |
Collapse
|
63
|
Pombo C, Wherry EJ, Gostick E, Price DA, Betts MR. Elevated Expression of CD160 and 2B4 Defines a Cytolytic HIV-Specific CD8+ T-Cell Population in Elite Controllers. J Infect Dis 2015; 212:1376-86. [PMID: 25883386 DOI: 10.1093/infdis/jiv226] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 04/06/2015] [Indexed: 01/30/2023] Open
Abstract
During chronic human immunodeficiency virus (HIV) infection, virus-specific CD8(+) T cells become functionally exhausted. Unlike most chronically infected individuals, elite controllers of HIV retain CD8(+) T-cell polyfunctionality and cytolytic capacity. It remains unclear whether elite controllers manifest T-cell exhaustion similar to subjects with chronic progression of HIV infection. Here we assessed coexpression of PD-1, Lag-3, CD160, and 2B4 as a measure of T-cell exhaustion in a cohort of elite controllers and in chronic progressors. We found that elite controllers have a high proportion of potentially exhausted (PD1(+)CD160(+)2B4(+)) HIV-specific CD8(+) T cells that is comparable to the proportion in chronic progressors. However, elite controllers also harbor a population of HIV-specific CD160(+)2B4(+) CD8(+) T cells that correlates with cytolytic capacity, as measured by perforin expression, a population not commonly present in chronic progressors. We therefore propose that coexpression of CD160 and 2B4 delineates a population of cytolytic CD8(+) T cells important for the control of HIV.
Collapse
Affiliation(s)
- Carolina Pombo
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - E John Wherry
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Emma Gostick
- Institute of Infection and Immunity, Cardiff University School of Medicine, United Kingdom
| | - David A Price
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland Institute of Infection and Immunity, Cardiff University School of Medicine, United Kingdom
| | - Michael R Betts
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
64
|
Šedý J, Bekiaris V, Ware CF. Tumor necrosis factor superfamily in innate immunity and inflammation. Cold Spring Harb Perspect Biol 2014; 7:a016279. [PMID: 25524549 DOI: 10.1101/cshperspect.a016279] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The tumor necrosis factor superfamily (TNFSF) and its corresponding receptor superfamily (TNFRSF) form communication pathways required for developmental, homeostatic, and stimulus-responsive processes in vivo. Although this receptor-ligand system operates between many different cell types and organ systems, many of these proteins play specific roles in immune system function. The TNFSF and TNFRSF proteins lymphotoxins, LIGHT (homologous to lymphotoxins, exhibits inducible expression, and competes with HSV glycoprotein D for herpes virus entry mediator [HVEM], a receptor expressed by T lymphocytes), lymphotoxin-β receptor (LT-βR), and HVEM are used by embryonic and adult innate lymphocytes to promote the development and homeostasis of lymphoid organs. Lymphotoxin-expressing innate-acting B cells construct microenvironments in lymphoid organs that restrict pathogen spread and initiate interferon defenses. Recent results illustrate how the communication networks formed among these cytokines and the coreceptors B and T lymphocyte attenuator (BTLA) and CD160 both inhibit and activate innate lymphoid cells (ILCs), innate γδ T cells, and natural killer (NK) cells. Understanding the role of TNFSF/TNFRSF and interacting proteins in innate cells will likely reveal avenues for future therapeutics for human disease.
Collapse
Affiliation(s)
- John Šedý
- Laboratory of Molecular Immunology, Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| | - Vasileios Bekiaris
- Laboratory of Molecular Immunology, Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| | - Carl F Ware
- Laboratory of Molecular Immunology, Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| |
Collapse
|
65
|
Licensed human natural killer cells aid dendritic cell maturation via TNFSF14/LIGHT. Proc Natl Acad Sci U S A 2014; 111:E5688-96. [PMID: 25512551 DOI: 10.1073/pnas.1411072112] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Interactions between natural killer (NK) cells and dendritic cells (DCs) aid DC maturation and promote T-cell responses. Here, we have analyzed the response of human NK cells to tumor cells, and we identify a pathway by which NK-DC interactions occur. Gene expression profiling of tumor-responsive NK cells identified the very rapid induction of TNF superfamily member 14 [TNFSF14; also known as homologous to lymphotoxins, exhibits inducible expression, and competes with HSV glycoprotein D for HVEM, a receptor expressed by T lymphocytes (LIGHT)], a cytokine implicated in the enhancement of antitumor responses. TNFSF14 protein expression was induced by three primary mechanisms of NK cell activation, namely, via the engagement of CD16, by the synergistic activity of multiple target cell-sensing NK-cell activation receptors, and by the cytokines IL-2 and IL-15. For antitumor responses, TNFSF14 was preferentially produced by the licensed NK-cell population, defined by the expression of inhibitory receptors specific for self-MHC class I molecules. In contrast, IL-2 and IL-15 treatment induced TNFSF14 production by both licensed and unlicensed NK cells, reflecting the ability of proinflammatory conditions to override the licensing mechanism. Importantly, both tumor- and cytokine-activated NK cells induced DC maturation in a TNFSF14-dependent manner. The coupling of TNFSF14 production to tumor-sensing NK-cell activation receptors links the tumor immune surveillance function of NK cells to DC maturation and adaptive immunity. Furthermore, regulation by NK cell licensing helps to safeguard against TNFSF14 production in response to healthy tissues.
Collapse
|
66
|
CD160 expression defines a uniquely exhausted subset of T lymphocytes in HTLV-1 infection. Biochem Biophys Res Commun 2014; 453:379-84. [PMID: 25277889 DOI: 10.1016/j.bbrc.2014.09.084] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 09/21/2014] [Indexed: 01/04/2023]
Abstract
HTLV-1 infection is a life-long retroviral infection. Chronic viral antigenic stimulation induces persistent infection which results in a clinically asymptomatic carrier state. Only a minor proportion of infected individuals develop adult T cell leukemia/lymphoma (ATLL) or HTLV-1-associated myelopathy/tropical spastic myelopathy (HAM/TSP). This is dependent on a balance of host and genetic factors. CD8+ cytotoxic T lymphocyte function is important in the immune response against viral infection; however, the contribution of CD160 receptor associated with CD8+ T lymphocytes is unclear. Thus, we sought to decipher its role on CTL function in HTLV-1 infection. Here, we report high frequencies of CD160 on CD8+ T cells, with significantly higher levels on HTLV-1 specific CD8+ T cells. Intercepting the CD160 pathway via blockade of the receptor or its ligand, herpes virus entry mediator (HVEM) resulted in improved perforin production and CD107a degranulation of HTLV-1 specific CD8+ T cells. Analysis of the CD160-expressing CD8+ cells demonstrated a unique subset associated with a highly differentiated effector memory based on CD45RA and CCR7 co-expression, increased expression of inhibitory molecules, 2B4 and PD1. Altogether, these results suggest a role for CD160/HVEM pathway in regulating immune response against HTLV-1 infection which may prove promising in the development of immune therapies for the treatment of HTLV-1 infection and other associated disorders.
Collapse
|
67
|
Sako N, Schiavon V, Bounfour T, Dessirier V, Ortonne N, Olive D, Ram-Wolff C, Michel L, Sicard H, Marie-Cardine A, Bagot M, Bensussan A, Schmitt C. Membrane expression of NK receptors CD160 and CD158k contributes to delineate a unique CD4+T-lymphocyte subset in normal and mycosis fungoides skin. Cytometry A 2014; 85:869-82. [DOI: 10.1002/cyto.a.22512] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 05/07/2014] [Accepted: 07/03/2014] [Indexed: 01/08/2023]
Affiliation(s)
- Nouhoum Sako
- INSERM; U976, F-75010 Paris France
- Univ Paris Diderot, Sorbonne Paris Cité; UMR-S 976, F-75739 Paris France
| | - Valérie Schiavon
- INSERM; U976, F-75010 Paris France
- Univ Paris Diderot, Sorbonne Paris Cité; UMR-S 976, F-75739 Paris France
| | - Touda Bounfour
- INSERM; U976, F-75010 Paris France
- Univ Paris Diderot, Sorbonne Paris Cité; UMR-S 976, F-75739 Paris France
| | - Valérie Dessirier
- INSERM; U976, F-75010 Paris France
- Univ Paris Diderot, Sorbonne Paris Cité; UMR-S 976, F-75739 Paris France
| | - Nicolas Ortonne
- Department of Pathology; AP-HP, Groupe Hospitalier Henri Mondor-Albert Chenevier, Henri Mondor Hospital; Créteil France
| | - Daniel Olive
- INSERM, UMR 891; Université de la Méditerranée, Institut Paoli Calmettes, Laboratoire d'Immunologie des Tumeurs; Marseille F-13009 France
| | - Caroline Ram-Wolff
- INSERM; U976, F-75010 Paris France
- Univ Paris Diderot, Sorbonne Paris Cité; UMR-S 976, F-75739 Paris France
- Dermatology Department; AP-HP, Hôp Saint Louis; F-75475 Paris France
| | - Laurence Michel
- INSERM; U976, F-75010 Paris France
- Univ Paris Diderot, Sorbonne Paris Cité; UMR-S 976, F-75739 Paris France
| | | | - Anne Marie-Cardine
- INSERM; U976, F-75010 Paris France
- Univ Paris Diderot, Sorbonne Paris Cité; UMR-S 976, F-75739 Paris France
| | - Martine Bagot
- INSERM; U976, F-75010 Paris France
- Univ Paris Diderot, Sorbonne Paris Cité; UMR-S 976, F-75739 Paris France
- Dermatology Department; AP-HP, Hôp Saint Louis; F-75475 Paris France
| | - Armand Bensussan
- INSERM; U976, F-75010 Paris France
- Univ Paris Diderot, Sorbonne Paris Cité; UMR-S 976, F-75739 Paris France
| | - Christian Schmitt
- INSERM; U976, F-75010 Paris France
- Univ Paris Diderot, Sorbonne Paris Cité; UMR-S 976, F-75739 Paris France
| |
Collapse
|
68
|
Gommerman JL, Browning JL, Ware CF. The Lymphotoxin Network: orchestrating a type I interferon response to optimize adaptive immunity. Cytokine Growth Factor Rev 2014; 25:139-45. [PMID: 24698108 DOI: 10.1016/j.cytogfr.2014.02.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Accepted: 02/24/2014] [Indexed: 12/13/2022]
Abstract
The Lymphotoxin (LT) pathway is best known for its role in orchestrating the development and homeostasis of lymph nodes and Peyer's patches through the regulation of homeostatic chemokines. More recently an appreciation of the LTβR pathway in the production of Type I interferons (IFN-I) during homeostasis and infection has emerged. LTβR signaling is essential in differentiating stromal cells and macrophages in lymphoid organs to rapidly produce IFN-I in response to virus infections independently of the conventional TLR signaling systems. In addition, LTβR signaling is required to produce homeostatic levels of IFN-I from dendritic cells in order to effectively cross-prime a CD8+ T cell response to protein antigen. Importantly, pharmacological inhibition of LTβR signaling in mice has a profound positive impact on a number of autoimmune disease models, although it remains unclear if this efficacy is linked to IFN-I production during chronic inflammation. In this review, we will provide a brief overview of how the "Lymphotoxin Network" is linked to the IFN-I response and its impact on the immune system.
Collapse
Affiliation(s)
| | - Jeffrey L Browning
- Department of Microbiology and Section of Rheumatology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Carl F Ware
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
69
|
Affiliation(s)
- Chris A Benedict
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA +1001 858 752 6732
| |
Collapse
|