1
|
Gogoleva VS, Drutskaya MS, Vorontsov AI, Atretkhany KSN, Belogurov AA, Kruglov AA, Nedospasov SA. Lymphotoxins from distinct types of lymphoid cells differentially contribute to neuroinflammation. Eur J Immunol 2024:e2350977. [PMID: 39210647 DOI: 10.1002/eji.202350977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Lymphotoxin α and lymphotoxin β (LTs), TNF superfamily members, are expressed in either soluble (LTα3) or membrane-bound (LTα1β2 or LTα2β1) forms. In the pathological context, LT-mediated signaling is known to exacerbate autoimmunity by perpetuating inflammation and promoting the formation of tertiary lymphoid organs. Despite this understanding, the exact roles of LTα and LTβ in the pathogenesis of the murine model of multiple sclerosis, and experimental autoimmune encephalomyelitis (EAE), remain controversial. Here, we employed a panel of gene-modified mice with cell-type restricted ablation of LTα (targeting both membrane-bound and soluble forms of LTs) to unravel the contributions of LTs from various lymphoid cells, namely T cells, type 3 innate lymphoid cells (ILC3) and B cells, in EAE. We found that the effects of LTα deletion were dependent on the cellular source. ILC3-derived lymphotoxins exerted a protective role in EAE by regulating the accumulation of IFN-ɣ- and GM-CSF-producing TH cells in the CNS. In contrast, T-cell-derived lymphotoxins promoted IL-17A- and GM-CSF-mediated TH responses in the periphery, whereas B-cell-derived lymphotoxins were pathogenic only in the autoantibody-mediated EAE model. Collectively, our findings unveil the multifaceted involvement of lymphotoxins in EAE pathogenesis and challenge the view that lymphotoxins play a solely pathogenic role in neuroinflammation.
Collapse
Affiliation(s)
- Violetta S Gogoleva
- Laboratory of Molecular Mechanisms of Immunity, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Moscow, 119991, Russia
| | - Marina S Drutskaya
- Laboratory of Molecular Mechanisms of Immunity, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Moscow, 119991, Russia
- Division of Immunobiology and Biomedicine, Sirius University of Science and Technology, Sirius, Krasnodarsky Krai, 354349, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Alexander I Vorontsov
- Laboratory of Molecular Mechanisms of Immunity, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Kamar-Sulu N Atretkhany
- Laboratory of Molecular Mechanisms of Immunity, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Alexey A Belogurov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Andrey A Kruglov
- AG Chronic Inflammation, German Rheumatism Research Center, a Leibniz Institute, Berlin, 10117, Germany
| | - Sergei A Nedospasov
- Laboratory of Molecular Mechanisms of Immunity, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Moscow, 119991, Russia
- Division of Immunobiology and Biomedicine, Sirius University of Science and Technology, Sirius, Krasnodarsky Krai, 354349, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| |
Collapse
|
2
|
Sukhumavasi W, Kaewamatawong T, Somboonpoonpol N, Jiratanh M, Wattanamethanont J, Kaewthamasorn M, Leelayoova S, Tiwananthagorn S. Liver- and Spleen-Specific Immune Responses in Experimental Leishmania martiniquensis Infection in BALB/c Mice. Front Vet Sci 2022; 8:794024. [PMID: 34977224 PMCID: PMC8718515 DOI: 10.3389/fvets.2021.794024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/24/2021] [Indexed: 11/13/2022] Open
Abstract
Leishmania martiniquensis is a neglected cause of an emerging leishmaniasis in many countries, including France, Germany, Switzerland, the United States of America, Myanmar, and Thailand, with different clinical manifestations ranging from asymptomatic, cutaneous (CL), visceral (VL), and atypically disseminated CL and VL. The persistence of parasites and the recurrence of the disease after treatment are challenges in controlling the disease. To explore efficient prophylaxis and therapy, this study aimed to investigate infection outcome and organ-specific immune responses after inoculation with L. martiniquensis (MHOM/TH/2011/PG; 5 x 106 promastigotes) in BALB/c mice via intravenous and intraperitoneal routes. A quantitative PCR technique, targeting L. martiniquensis ITS1, was primarily established to estimate the parasite burden. We found that the infection in the liver resolved; however, persistent infection was observed in the spleen. Histopathology with Leishmania-specific immunostaining revealed efficient hepatic granuloma formation, while splenic disorganization with parasitized macrophages at different locations was demonstrated. The mRNA expression of Th1 cytokines (IFN-γ, TNF-α, IL-12p40) and iNOS in the liver and spleen was upregulated. In addition, high expression of IL-10 was observed in the spleen in the chronic phase, revealing a significant moderate correlation with the parasite persistence [r(12) = 0.72, P = 0.009]. Further clarification of the mechanisms of persistent infection and experimental infection in immunosuppressed murine models are warranted.
Collapse
Affiliation(s)
- Woraporn Sukhumavasi
- Parasitology Unit, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.,Feline Infectious Disease and Health for Excellence Research Unit, Microbial Food Safety and Antimicrobial Resistance Research Unit, Animal Vector-Borne Disease Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Theerayuth Kaewamatawong
- Veterinary Pathology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Nawaphat Somboonpoonpol
- Parasitology Unit, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Montakan Jiratanh
- Parasitology Section, National Institute of Animal Health, Department of Livestock Development, Ministry of Agriculture and Cooperatives, Bangkok, Thailand
| | - Juntra Wattanamethanont
- Parasitology Section, National Institute of Animal Health, Department of Livestock Development, Ministry of Agriculture and Cooperatives, Bangkok, Thailand
| | - Morakot Kaewthamasorn
- Parasitology Unit, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.,Veterinary Parasitology Research Unit, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Saovanee Leelayoova
- Department of Parasitology, Phramongkutklao College of Medicine, Bangkok, Thailand
| | - Saruda Tiwananthagorn
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand.,Research Center of Producing and Development of Products and Innovations for Animal Health and Production, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
3
|
Vyas AK, Lslam M, Garg G, Singh AK, Trehanpati N. Humoral Immune Responses and Hepatitis B Infection. Dig Dis 2021; 39:516-525. [PMID: 33429386 DOI: 10.1159/000514274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/11/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Chronicity or seroclearance of hepatitis B virus (HBV) antigens is determined by the host immune responses. Current approaches to treat HBV patients are based on inhibition of replication using different antivirals (nucleoside or nucleotide analogs) as monotherapy, or along with immune modulators as combination therapy is being used worldwide for reducing the viral load. Understanding the role of immune cellular therapies with currently available treatments for persistent viral-mediated responses in HBV patients is unexplored. However, the generation of antibodies against a surface (HBs) and envelop (HBe) antigen of hepatitis B remains an issue for future studies and needs to be explored. SUMMARY Humoral immunity, specifically T follicular helper (TFh) cells, may serve as a target for therapy for HBsAg seroconversion. In this review, we have been engrossed in the importance and role of the humoral immune responses in CHBV infection and vertical transmission. Key Message: TFh cells have been suggested as the potential target of immunotherapy which lead to seroconversion of HBe and HBs antigens of HBV. HBsAg seroconversion and eradication of covalently closed circular DNA are the main challenges for existing and forthcoming therapies in HBV infection.
Collapse
Affiliation(s)
- Ashish Kumar Vyas
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal, India
| | - Mojahidul Lslam
- Departments of Molecular and Cellular Medicine, Institute of Liver & Biliary Sciences, New Delhi, India
| | - Garima Garg
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal, India
| | - Anirudh K Singh
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal, India
| | - Nirupma Trehanpati
- Departments of Molecular and Cellular Medicine, Institute of Liver & Biliary Sciences, New Delhi, India
| |
Collapse
|
4
|
Mafra K, Nakagaki BN, Castro Oliveira HM, Rezende RM, Antunes MM, Menezes GB. The liver as a nursery for leukocytes. J Leukoc Biol 2019; 106:687-693. [PMID: 31107980 DOI: 10.1002/jlb.mr1118-455r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/10/2019] [Accepted: 04/26/2019] [Indexed: 12/20/2022] Open
Abstract
Leukocytes are a large population of cells spread within most tissues in the body. These cells may be either sessile (called as resident cells) or circulating leukocytes, which travel long journeys inside the vessels during their lifespan. Although production and maturation of these leukocytes in adults primarily occur in the bone marrow, it is well known that this process-called hematopoiesis-started in the embryonic life in different sites, including the yolk sac, placenta, and the liver. In this review, we will discuss how the liver acts as a pivotal site for leukocyte maturation during the embryo phase, and also how the most frequent liver-resident immune cell populations-namely Kupffer cells, dendritic cells, and lymphocytes-play a vital role in both tolerance and inflammatory responses to antigens from food, microbiota, and pathogens.
Collapse
Affiliation(s)
- Kassiana Mafra
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Brenda Naemi Nakagaki
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Hortência Maciel Castro Oliveira
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rafael Machado Rezende
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Maísa Mota Antunes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gustavo Batista Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
5
|
Louveau A, Da Mesquita S, Kipnis J. Lymphatics in Neurological Disorders: A Neuro-Lympho-Vascular Component of Multiple Sclerosis and Alzheimer's Disease? Neuron 2016; 91:957-973. [PMID: 27608759 PMCID: PMC5019121 DOI: 10.1016/j.neuron.2016.08.027] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lymphatic vasculature drains interstitial fluids, which contain the tissue's waste products, and ensures immune surveillance of the tissues, allowing immune cell recirculation. Until recently, the CNS was considered to be devoid of a conventional lymphatic vasculature. The recent discovery in the meninges of a lymphatic network that drains the CNS calls into question classic models for the drainage of macromolecules and immune cells from the CNS. In the context of neurological disorders, the presence of a lymphatic system draining the CNS potentially offers a new player and a new avenue for therapy. In this review, we will attempt to integrate the known primary functions of the tissue lymphatic vasculature that exists in peripheral organs with the proposed function of meningeal lymphatic vessels in neurological disorders, specifically multiple sclerosis and Alzheimer's disease. We propose that these (and potentially other) neurological afflictions can be viewed as diseases with a neuro-lympho-vascular component and should be therapeutically targeted as such.
Collapse
Affiliation(s)
- Antoine Louveau
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Sandro Da Mesquita
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
6
|
Becher B. The Good, the Bad, or the Pretty: IL-17 Builds Lymphoid Tissues in the Brain. Immunity 2016; 43:1033-4. [PMID: 26682978 DOI: 10.1016/j.immuni.2015.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Lymph node development depends on well-orchestrated interactions between lymphoid tissue inducer cells and stromal cells. In this issue of Immunity, Pikor and colleagues (2015) find that signals from IL-17-producing T helper cells can alter the stromal microenvironment of the inflamed brain to generate a neo-lymphoid organ that drives further inflammation.
Collapse
Affiliation(s)
- Burkhard Becher
- Institute of Experimental Immunology, Inflammation Research, University of Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
7
|
Thomas SN, Rohner NA, Edwards EE. Implications of Lymphatic Transport to Lymph Nodes in Immunity and Immunotherapy. Annu Rev Biomed Eng 2016; 18:207-33. [PMID: 26928210 DOI: 10.1146/annurev-bioeng-101515-014413] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Adaptive immune response consists of many highly regulated, multistep cascades that protect against infection while preserving the health of autologous tissue. The proper initiation, maintenance, and resolution of such responses require the precise coordination of molecular and cellular signaling over multiple time and length scales orchestrated by lymphatic transport. In order to investigate these functions and manipulate them for therapy, a comprehensive understanding of how lymphatics influence immune physiology is needed. This review presents the current mechanistic understanding of the role of the lymphatic vasculature in regulating biomolecule and cellular transport from the interstitium, peripheral tissue immune surveillance, the lymph node stroma and microvasculature, and circulating lymphocyte homing to lymph nodes. This review also discusses the ramifications of lymphatic transport in immunity as well as tolerance and concludes with examples of how lymphatic-mediated targeting of lymph nodes has been exploited for immunotherapy applications.
Collapse
Affiliation(s)
- Susan N Thomas
- George W. Woodruff School of Mechanical Engineering and.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332; .,Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Nathan A Rohner
- George W. Woodruff School of Mechanical Engineering and.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332;
| | - Erin E Edwards
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332; .,Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332
| |
Collapse
|
8
|
Pikor N, Astarita J, Summers-Deluca L, Galicia G, Qu J, Ward L, Armstrong S, Dominguez C, Malhotra D, Heiden B, Kay R, Castanov V, Touil H, Boon L, O’Connor P, Bar-Or A, Prat A, Ramaglia V, Ludwin S, Turley S, Gommerman J. Integration of Th17- and Lymphotoxin-Derived Signals Initiates Meningeal-Resident Stromal Cell Remodeling to Propagate Neuroinflammation. Immunity 2015; 43:1160-73. [DOI: 10.1016/j.immuni.2015.11.010] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 06/22/2015] [Accepted: 09/08/2015] [Indexed: 12/16/2022]
|
9
|
Katakam AK, Brightbill H, Franci C, Kung C, Nunez V, Jones C, Peng I, Jeet S, Wu LC, Mellman I, Delamarre L, Austin CD. Dendritic cells require NIK for CD40-dependent cross-priming of CD8+ T cells. Proc Natl Acad Sci U S A 2015; 112:14664-9. [PMID: 26561586 PMCID: PMC4664370 DOI: 10.1073/pnas.1520627112] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Dendritic cells (DCs) link innate and adaptive immunity and use a host of innate immune and inflammatory receptors to respond to pathogens and inflammatory stimuli. Although DC maturation via canonical NF-κB signaling is critical for many of these functions, the role of noncanonical NF-κB signaling via the serine/threonine kinase NIK (NF-κB-inducing kinase) remains unclear. Because NIK-deficient mice lack secondary lymphoid organs, we generated transgenic mice with targeted NIK deletion in CD11c(+) cells. Although these mice exhibited normal lymphoid organs, they were defective in cross-priming naive CD8(+) T cells following vaccination, even in the presence of anti-CD40 or polyinosinic:polycytidylic acid to induce DC maturation. This impairment reflected two intrinsic defects observed in splenic CD8(+) DCs in vitro, namely antigen cross-presentation to CD8(+) T cells and secretion of IL-12p40, a cytokine known to promote cross-priming in vivo. In contrast, antigen presentation to CD4(+) T cells was not affected. These findings reveal that NIK, and thus probably the noncanonical NF-κB pathway, is critical to allow DCs to acquire the capacity to cross-present antigen and prime CD8 T cells after exposure to licensing stimuli, such as an agonistic anti-CD40 antibody or Toll-like receptor 3 ligand.
Collapse
Affiliation(s)
- Anand K Katakam
- Department of Pathology, Genentech Inc., South San Francisco, CA 94080
| | - Hans Brightbill
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080
| | - Christian Franci
- Department of Cancer Immunology, Genentech Inc., South San Francisco, CA 94080
| | - Chung Kung
- Department of Mouse Genetics, Genentech Inc., South San Francisco, CA 94080
| | - Victor Nunez
- Department of Pathology, Genentech Inc., South San Francisco, CA 94080
| | - Charles Jones
- Department of Pathology, Genentech Inc., South San Francisco, CA 94080
| | - Ivan Peng
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080
| | - Surinder Jeet
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080
| | - Lawren C Wu
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080
| | - Ira Mellman
- Department of Cancer Immunology, Genentech Inc., South San Francisco, CA 94080;
| | - Lélia Delamarre
- Department of Cancer Immunology, Genentech Inc., South San Francisco, CA 94080
| | - Cary D Austin
- Department of Pathology, Genentech Inc., South San Francisco, CA 94080;
| |
Collapse
|
10
|
Group 3 innate lymphoid cells accumulate and exhibit disease-induced activation in the meninges in EAE. Cell Immunol 2015; 297:69-79. [PMID: 26163773 DOI: 10.1016/j.cellimm.2015.06.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 05/31/2015] [Accepted: 06/18/2015] [Indexed: 12/11/2022]
Abstract
Innate lymphoid cells are immune cells that reside in tissues that interface with the external environment and contribute to the first line defense against pathogens. However, they also have roles in promoting chronic inflammation. Here we demonstrate that group 3 ILCs, (ILC3s - CD45+Lin-IL-7Rα+RORγt+), are normal residents of the meninges and exhibit disease-induced accumulation and activation in EAE. In addition to production of the pro-inflammatory cytokines IL-17 and GM-CSF, ILC3s constitutively express CD30L and OX40L, molecules required for memory T cell survival. We show that disease-induced trafficking of transferred wild type T cells to the meninges is impaired in ILC3-deficient Rorc-/- mice. Furthermore, lymphoid tissue inducer cells, a c-kit+ ILC3 subset that promotes ectopic lymphoid follicle development, a hallmark of many autoimmune diseases, are reduced in the meninges of EAE-resistant c-kit mutant Kit(W/Wv) mice. We propose that ILC3s sustain neuroinflammation by supporting T cell survival and reactivation in the meninges.
Collapse
|
11
|
Kolesnikov SI, Michurina SV, Arkhipov SA. Changes in Peripheral Blood Monocytes and Liver Macrophages in Male Rats after Benzo(a)pyrene Injection. Bull Exp Biol Med 2015; 158:735-8. [PMID: 25894771 DOI: 10.1007/s10517-015-2850-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Indexed: 11/28/2022]
Abstract
Intraperitoneal injections of benzo(a)pyrene to male rats in a total dose of 60 mg/kg modified the production of ROS and the phagocytic potential of blood monocytes by modulating their potential bactericidal activity. The lysosomal system (particularly the secondary lysosomes) of liver macrophages was activated, which promoted fusion of the hydrolytic potentials of macrophages and monocytes. These results indicated that the toxin modulated the cellular immune homeostasis and the level of general nonspecific resistance.
Collapse
Affiliation(s)
- S I Kolesnikov
- Research Center of Family Health Problems and Human Reproduction, Irkutsk, Russia,
| | | | | |
Collapse
|
12
|
Abstract
Gold Standard allergen-specific immunotherapy is associated with low efficacy because it requires either many subcutaneous injections of allergen or even more numerous sublingual allergen administrations to achieve amelioration of symptoms. Intralymphatic vaccination can maximize immunogenicity and hence efficacy. We and others have demonstrated that as few as three low dose intralymphatic allergen administrations are sufficient to effectively alleviate symptoms. Results of recent prospective and controlled trials suggest that this strategy may be an effective form of allergen immunotherapy.
Collapse
Affiliation(s)
- Gabriela Senti
- Clinical Trials Center, University Hospital Zurich, Raemistrasse 100/MOU2, CH-8091 Zurich, Switzerland
| | - Thomas M Kündig
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Li Y, Tang L, Hou J. Role of interleukin-21 in HBV infection: friend or foe? Cell Mol Immunol 2014; 12:303-8. [PMID: 25363528 DOI: 10.1038/cmi.2014.109] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 09/23/2014] [Accepted: 10/06/2014] [Indexed: 12/20/2022] Open
Abstract
Interleukin (IL)-21, a cytokine produced by activated CD4(+) T cells, has broad pleiotropic actions that affect the functions of a variety of lymphoid cells. The roles of IL-21 in modulating immunity to infections are currently being defined. Notably, IL-21-mediated cellular and humoral immune responses play an important role in determining the outcome of viral infection. This article reviews the current knowledge on the critical role of IL-21 in hepatitis B virus (HBV) infection. As a competent intermediary, IL-21 derived from virus-specific CD4(+) T cells plays key roles in sustaining CD8(+) T cells and promoting B-cell responses that are essential for effective viral control. However, as a mediator of inflammation, IL-21 is also involved in the development of HBV-induced liver cirrhosis and exacerbating liver injury. Overall, the current data point to IL-21 as an immunomodulatory cytokine in HBV infection. Immunotherapeutic strategies aimed at optimizing the beneficial effects of IL-21 in HBV infection may prove to be a rigorous challenge in the future, as they should foster the strengths of IL-21 while circumventing potential drawbacks.
Collapse
Affiliation(s)
- Yongyin Li
- 1] Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China [3] Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| | - Libo Tang
- 1] Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Jinlin Hou
- 1] Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, China [2] State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China [3] Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| |
Collapse
|
14
|
Bunn PT, Stanley AC, de Labastida Rivera F, Mulherin A, Sheel M, Alexander CE, Faleiro RJ, Amante FH, Montes De Oca M, Best SE, James KR, Kaye PM, Haque A, Engwerda CR. Tissue requirements for establishing long-term CD4+ T cell-mediated immunity following Leishmania donovani infection. THE JOURNAL OF IMMUNOLOGY 2014; 192:3709-18. [PMID: 24634490 DOI: 10.4049/jimmunol.1300768] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Organ-specific immunity is a feature of many infectious diseases, including visceral leishmaniasis caused by Leishmania donovani. Experimental visceral leishmaniasis in genetically susceptible mice is characterized by an acute, resolving infection in the liver and chronic infection in the spleen. CD4+ T cell responses are critical for the establishment and maintenance of hepatic immunity in this disease model, but their role in chronically infected spleens remains unclear. In this study, we show that dendritic cells are critical for CD4+ T cell activation and expansion in all tissue sites examined. We found that FTY720-mediated blockade of T cell trafficking early in infection prevented Ag-specific CD4+ T cells from appearing in lymph nodes, but not the spleen and liver, suggesting that early CD4+ T cell priming does not occur in liver-draining lymph nodes. Extended treatment with FTY720 over the first month of infection increased parasite burdens, although this associated with blockade of lymphocyte egress from secondary lymphoid tissue, as well as with more generalized splenic lymphopenia. Importantly, we demonstrate that CD4+ T cells are required for the establishment and maintenance of antiparasitic immunity in the liver, as well as for immune surveillance and suppression of parasite outgrowth in chronically infected spleens. Finally, although early CD4+ T cell priming appeared to occur most effectively in the spleen, we unexpectedly revealed that protective CD4+ T cell-mediated hepatic immunity could be generated in the complete absence of all secondary lymphoid tissues.
Collapse
Affiliation(s)
- Patrick T Bunn
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Publicover J, Gaggar A, Nishimura S, Van Horn CM, Goodsell A, Muench MO, Reinhardt RL, van Rooijen N, Wakil AE, Peters M, Cyster JG, Erle DJ, Rosenthal P, Cooper S, Baron JL. Age-dependent hepatic lymphoid organization directs successful immunity to hepatitis B. J Clin Invest 2013; 123:3728-39. [PMID: 23925290 DOI: 10.1172/jci68182] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 06/06/2013] [Indexed: 02/06/2023] Open
Abstract
Hepatitis B virus (HBV) is a major human pathogen that causes immune-mediated hepatitis. Successful immunity to HBV is age dependent: viral clearance occurs in most adults, whereas neonates and young children usually develop chronic infection. Using a mouse model of HBV infection, we sought mechanisms underpinning the age-dependent outcome of HBV and demonstrated that hepatic macrophages facilitate lymphoid organization and immune priming within the adult liver and promote successful immunity. In contrast, lymphoid organization and immune priming was greatly diminished in the livers of young mice, and of macrophage-depleted adult mice, leading to abrogated HBV immunity. Furthermore, we found that CXCL13, which is involved in B lymphocyte trafficking and lymphoid architecture and development, is expressed in an age-dependent manner in both adult mouse and human hepatic macrophages and plays an integral role in facilitating an effective immune response against HBV. Taken together, these results identify some of the immunological mechanisms necessary for effective control of HBV.
Collapse
Affiliation(s)
- Jean Publicover
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Communication between pathogenic T cells and myeloid cells in neuroinflammatory disease. Trends Immunol 2013; 34:114-9. [DOI: 10.1016/j.it.2012.09.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 09/17/2012] [Accepted: 09/27/2012] [Indexed: 12/23/2022]
|
17
|
Becher B, Greter M. Acquitting an APC: DCs found “not guilty” after trial by ablation. Eur J Immunol 2012; 42:2551-4. [DOI: 10.1002/eji.201242928] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Burkhard Becher
- Institute of Experimental Immunology; University of Zurich; Zurich; Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology; University of Zurich; Zurich; Switzerland
| |
Collapse
|
18
|
Tiwananthagorn S, Iwabuchi K, Ato M, Sakurai T, Kato H, Katakura K. Involvement of CD4⁺ Foxp3⁺ regulatory T cells in persistence of Leishmania donovani in the liver of alymphoplastic aly/aly mice. PLoS Negl Trop Dis 2012; 6:e1798. [PMID: 22928057 PMCID: PMC3424244 DOI: 10.1371/journal.pntd.0001798] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 07/16/2012] [Indexed: 11/17/2022] Open
Abstract
Visceral leishmaniasis (VL) is a chronic and fatal disease in humans and dogs caused by the intracellular protozoan parasites, Leishmania donovani and L. infantum (L. chagasi). Relapse of disease is frequent in immunocompromised patients, in which the number of VL cases has been increasing recently. The present study is aimed to improve the understanding of mechanisms of L. donovani persistence in immunocompromised conditions using alymphoplastic aly/aly mice. Hepatic parasite burden, granuloma formation and induction of regulatory T cells were determined for up to 7 months after the intravenous inoculation with L. donovani promastigotes. While control aly/+ mice showed a peak of hepatic parasite growth at 4 weeks post infection (WPI) and resolved the infection by 8 WPI, aly/aly mice showed a similar peak in hepatic parasite burden but maintained persistent in the chronic phase of infection, which was associated with delayed and impaired granuloma maturation. Although hepatic CD4+Foxp3+ but not CD8+Foxp3+ T cells were first detected at 4 WPI in both strains of mice, the number of CD4+Foxp3+ T cells was significantly increased in aly/aly mice from 8 WPI. Immunohistochemical analysis demonstrated the presence of Foxp3+ T cells in L. donovani–induced hepatic granulomas and perivascular neo-lymphoid aggregates. Quantitative real-time PCR analysis of mature granulomas collected by laser microdissection revealed the correlation of Foxp3 and IL-10 mRNA level. Furthermore, treatment of infected aly/aly mice with anti-CD25 or anti-FR4 mAb resulted in significant reductions in both hepatic Foxp3+ cells and parasite burden. Thus, we provide the first evidence that CD4+Foxp3+ Tregs mediate L. donovani persistence in the liver during VL in immunodeficient murine model, a result that will help to establish new strategies of immunotherapy against this intracellular protozoan pathogen. The protozoan parasite Leishmania donovani is the causative agent of visceral leishmaniasis (VL) with a variety of outcomes ranging from asymptomatic to fatal infection. In the last decade, an increasing number of VL cases in immunocompromised conditions have been reported. Loss of the control of parasite persistence causes relapse of the disease in these patients. To clarify why parasite persistence and disease are caused in an immunocompromised condition, we examined L. donovani infection in alymphoplastic aly/aly mice that completely lack lymph nodes and have disturbed spleen architecture. Although parasites grew in the liver of aly/+ mice for the first 4 weeks post infection (WPI) and parasites were eliminated by 8 WPI, we found that parasites persisted in the liver of aly/aly mice with the ineffective of granuloma formation to kill the parasites. These aly/aly mice showed significant increases in CD4+Foxp3+ regulatory T cells in the liver. Consequently, we treated infected mice with anti-CD25 or anti-FR4 mAb to inhibit the function of Tregs, and found significant reductions in both hepatic Foxp3+ cells and parasite burden. These results clearly demonstrated for the first time that the expansion of CD4+Foxp3+ Tregs is involved in hepatic L. donovani persistence in immunodeficient murine model.
Collapse
Affiliation(s)
- Saruda Tiwananthagorn
- Laboratory of Parasitology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | |
Collapse
|
19
|
Thomas SN, Rutkowski JM, Pasquier M, Kuan EL, Alitalo K, Randolph GJ, Swartz MA. Impaired humoral immunity and tolerance in K14-VEGFR-3-Ig mice that lack dermal lymphatic drainage. THE JOURNAL OF IMMUNOLOGY 2012; 189:2181-90. [PMID: 22844119 DOI: 10.4049/jimmunol.1103545] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Lymphatic vessels transport interstitial fluid, soluble Ag, and immune cells from peripheral tissues to lymph nodes (LNs), yet the contribution of peripheral lymphatic drainage to adaptive immunity remains poorly understood. We examined immune responses to dermal vaccination and contact hypersensitivity (CHS) challenge in K14-VEGFR-3-Ig mice, which lack dermal lymphatic capillaries and experience markedly depressed transport of solutes and dendritic cells from the skin to draining LNs. In response to dermal immunization, K14-VEGFR-3-Ig mice produced lower Ab titers. In contrast, although delayed, T cell responses were robust after 21 d, including high levels of Ag-specific CD8+ T cells and production of IFN-γ, IL-4, and IL-10 upon restimulation. T cell-mediated CHS responses were strong in K14-VEGFR-3-Ig mice, but importantly, their ability to induce CHS tolerance in the skin was impaired. In addition, 1-y-old mice displayed multiple signs of autoimmunity. These data suggest that lymphatic drainage plays more important roles in regulating humoral immunity and peripheral tolerance than in effector T cell immunity.
Collapse
Affiliation(s)
- Susan N Thomas
- Institute of Bioengineering, School of Life Sciences, Swiss Federal Institute of Technology, Lausanne-EPFL, 1015 Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
20
|
Häcker H, Chi L, Rehg JE, Redecke V. NIK prevents the development of hypereosinophilic syndrome-like disease in mice independent of IKKα activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2012; 188:4602-10. [PMID: 22474019 PMCID: PMC3532048 DOI: 10.4049/jimmunol.1200021] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Immune cell-mediated tissue injury is a common feature of different inflammatory diseases, yet the pathogenetic mechanisms and cell types involved vary significantly. Hypereosinophilic syndrome (HES) represents a group of inflammatory diseases that is characterized by increased numbers of pathogenic eosinophilic granulocytes in the peripheral blood and diverse organs. On the basis of clinical and laboratory findings, various forms of HES have been defined, yet the molecular mechanism and potential signaling pathways that drive eosinophil expansion remain largely unknown. In this study, we show that mice deficient of the serine/threonine-specific protein kinase NF-κB-inducing kinase (NIK) develop a HES-like disease, reflected by progressive blood and tissue eosinophilia, tissue injury, and premature death at around 25-30 wk of age. Similar to the lymphocytic form of HES, CD4(+) T cells from NIK-deficient mice express increased levels of Th2-associated cytokines, and eosinophilia and survival of NIK-deficient mice could be prevented completely by genetic ablation of CD4(+) T cells. Experiments based on bone marrow chimeric mice, however, demonstrated that inflammation in NIK-deficient mice depended on radiation-resistant tissues, implicating that NIK-deficient immune cells mediate inflammation in a nonautonomous manner. Surprisingly, disease development was independent of NIK's known function as an IκB kinase α (IKKα) kinase, because mice carrying a mutation in the activation loop of IKKα, which is phosphorylated by NIK, did not develop inflammatory disease. Our data show that NIK activity in nonhematopoietic cells controls Th2 cell development and prevents eosinophil-driven inflammatory disease, most likely using a signaling pathway that operates independent of the known NIK substrate IKKα.
Collapse
Affiliation(s)
- Hans Häcker
- Department of Infectious Diseases, St Jude Children’s Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105, USA
| | - Liying Chi
- Department of Infectious Diseases, St Jude Children’s Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105, USA
| | - Jerold E. Rehg
- Department of Pathology, St Jude Children’s Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105, USA
| | - Vanessa Redecke
- Department of Infectious Diseases, St Jude Children’s Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105, USA
| |
Collapse
|
21
|
Moore JWJ, Beattie L, Dalton JE, Owens BMJ, Maroof A, Coles MC, Kaye PM. B cell: T cell interactions occur within hepatic granulomas during experimental visceral leishmaniasis. PLoS One 2012; 7:e34143. [PMID: 22479545 PMCID: PMC3316612 DOI: 10.1371/journal.pone.0034143] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 02/22/2012] [Indexed: 11/18/2022] Open
Abstract
Hepatic resistance to Leishmania donovani infection in mice is associated with the development of granulomas, in which a variety of lymphoid and non-lymphoid populations accumulate. Although previous studies have identified B cells in hepatic granulomas and functional studies in B cell-deficient mice have suggested a role for B cells in the control of experimental visceral leishmaniasis, little is known about the behaviour of B cells in the granuloma microenvironment. Here, we first compared the hepatic B cell population in infected mice, where ≈60% of B cells are located within granulomas, with that of naïve mice. In infected mice, there was a small increase in mIgM(lo)mIgD(+) mature B2 cells, but no enrichment of B cells with regulatory phenotype or function compared to the naïve hepatic B cell population, as assessed by CD1d and CD5 expression and by IL-10 production. Using 2-photon microscopy to quantify the entire intra-granuloma B cell population, in conjunction with the adoptive transfer of polyclonal and HEL-specific BCR-transgenic B cells isolated from L. donovani-infected mice, we demonstrated that B cells accumulate in granulomas over time in an antigen-independent manner. Intra-vital dynamic imaging was used to demonstrate that within the polyclonal B cell population obtained from L. donovani-infected mice, the frequency of B cells that made multiple long contacts with endogenous T cells was greater than that observed using HEL-specific B cells obtained from the same inflammatory environment. These data indicate, therefore, that a subset of this polyclonal B cell population is capable of making cognate interactions with T cells within this unique environment, and provide the first insights into the dynamics of B cells within an inflammatory site.
Collapse
Affiliation(s)
- John W. J. Moore
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, Heslington, York, United Kingdom
| | - Lynette Beattie
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, Heslington, York, United Kingdom
| | - Jane E. Dalton
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, Heslington, York, United Kingdom
| | - Benjamin M. J. Owens
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, Heslington, York, United Kingdom
| | - Asher Maroof
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, Heslington, York, United Kingdom
| | - Mark C. Coles
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, Heslington, York, United Kingdom
| | - Paul M. Kaye
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, Heslington, York, United Kingdom
| |
Collapse
|
22
|
Hofmann J, Mair F, Greter M, Schmidt-Supprian M, Becher B. NIK signaling in dendritic cells but not in T cells is required for the development of effector T cells and cell-mediated immune responses. J Exp Med 2011; 208:1917-29. [PMID: 21807870 PMCID: PMC3171087 DOI: 10.1084/jem.20110128] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 06/29/2011] [Indexed: 12/24/2022] Open
Abstract
The canonical NF-κB pathway is a driving force for virtually all aspects of inflammation. Conversely, the role of the noncanonical NF-κB pathway and its central mediator NF-κB-inducing kinase (NIK) remains poorly defined. NIK has been proposed to be involved in the formation of T(H)17 cells, and its absence in T(H) cells renders them incapable of inducing autoimmune responses, suggesting a T cell-intrinsic role for NIK. Upon systematic analysis of NIK function in cell-mediated immunity, we found that NIK signaling is dispensable within CD4(+) T cells but played a pivotal role in dendritic cells (DCs). We discovered that NIK signaling is required in DCs to deliver co-stimulatory signals to CD4(+) T cells and that DC-restricted expression of NIK is sufficient to restore T(H)1 and T(H)17 responses as well as cell-mediated immunity in NIK(-/-) mice. When CD4(+) T cells developed in the absence of NIK-sufficient DCs, they were rendered anergic. Reintroduction of NIK into DCs allowed developing NIK(-/-) CD4(+) T cells to become functional effector populations and restored the development of autoimmune disease. Therefore, our data suggest that a population of thymic DCs requires NIK to shape the formation of most αβ CD4(+) T effector lineages during early development.
Collapse
MESH Headings
- Animals
- Clonal Anergy/genetics
- Clonal Anergy/immunology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Immunity, Cellular/physiology
- Mice
- Mice, Knockout
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/immunology
- Protein Serine-Threonine Kinases/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Signal Transduction/immunology
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th17 Cells/immunology
- Th17 Cells/metabolism
- NF-kappaB-Inducing Kinase
Collapse
Affiliation(s)
- Janin Hofmann
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Florian Mair
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Marc Schmidt-Supprian
- Molecular Immunology and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
23
|
Manifestation of spontaneous and early autoimmune gastritis in CCR7-deficient mice. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:754-65. [PMID: 21801869 DOI: 10.1016/j.ajpath.2011.04.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 03/16/2011] [Accepted: 04/12/2011] [Indexed: 11/20/2022]
Abstract
Autoimmune gastritis is a common autoimmune disorder characterized by chronic inflammatory cell infiltrates, atrophy of the corpus and fundus, and the occurrence of autoantibodies to parietal cell antigen. In CCR7-deficient mice, autoimmune gastritis developed spontaneously and was accompanied by metaplasia of the gastric mucosa and by the formation of tertiary lymphoid organs at gastric mucosal sites. T cells of CCR7-deficient mice showed an activated phenotype in the gastric mucosa, mesenteric lymph nodes, and peripheral blood. In addition, elevated serum IgG levels specific to gastric parietal cell antigen were detected. Because the role of organized lymphocytic aggregates at this inflammatory site is not completely understood, we first analyzed the cellular requirements for the formation of these structures. Autoreactive CD4(+) T cells were pivotal for tertiary lymphoid follicle formation, most likely in cooperation with dendritic cells, macrophages, and B cells. Second, we analyzed the necessity of secondary lymph nodes and tertiary lymphoid organs for the development of autoimmune gastritis using CCR7 single- and CCR7/lymphotoxin α double-deficient mice. Strikingly, manifestation of autoimmune gastritis was observed in the absence of secondary lymph nodes and preceded the development of tertiary lymphoid organs. Taken together, these findings identify an inflammatory process where gastric autoreactive T cells independent of organized tertiary lymphoid organs and classic lymph nodes can induce and maintain autoimmune gastritis.
Collapse
|
24
|
Siegmund K, Zeis T, Kunz G, Rolink T, Schaeren-Wiemers N, Pieters J. Coronin 1-Mediated Naive T Cell Survival Is Essential for the Development of Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2011; 186:3452-61. [DOI: 10.4049/jimmunol.1003491] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
25
|
Senti G, Johansen P, Kündig TM. Intralymphatic immunotherapy: from the rationale to human applications. Curr Top Microbiol Immunol 2011; 352:71-84. [PMID: 21725898 DOI: 10.1007/82_2011_133] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Allergen specific immunotherapy (SIT) is the only treatment of IgE mediated allergies that is causative and has a long-term effect. Classically, SIT requires numerous subcutaneous injections of the allergen during 3-5 years. Over the last decade sublingual allergen applications have established as an alternative, but treatment duration could not be shortened. This review focuses on direct administration of vaccines in general and of allergens in particular into lymph nodes with the aim to enhance immunotherapy. Several studies have found that direct injection of antigens into lymph nodes enhanced immune responses. Recently we have focused on intralymphatic allergen administration in order to enhance SIT. Data in mouse models and in clinical trials showed that intralymphatic allergen administration strongly enhanced SIT, so that the number of allergen injections could be reduced to three, and the allergen dose could be reduced 10-100 fold. Intralymphatic injections proved easy, practically painless and safe. In mice and men, intralymphatic immunotherapy injecting allergens into a subcutaneous lymph node markedly enhances the protective immune response, so that both the dose and number of allergen injections can be reduced, making SIT safer and faster, which enhances patient convenience and compliance.
Collapse
Affiliation(s)
- Gabriela Senti
- Clinical Trials Center, Center for Clinical Research, University and University Hospital of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland.
| | | | | |
Collapse
|
26
|
Harp JR, Onami TM. Naïve T cells re-distribute to the lungs of selectin ligand deficient mice. PLoS One 2010; 5:e10973. [PMID: 20532047 PMCID: PMC2881108 DOI: 10.1371/journal.pone.0010973] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 05/07/2010] [Indexed: 11/18/2022] Open
Abstract
Background Selectin mediated tethering represents one of the earliest steps in T cell extravasation into lymph nodes via high endothelial venules and is dependent on the biosynthesis of sialyl Lewis X (sLex) ligands by several glycosyltransferases, including two fucosyltransferases, fucosyltransferase-IV and –VII. Selectin mediated binding also plays a key role in T cell entry to inflamed organs. Methodology/Principal Findings To understand how loss of selectin ligands (sLex) influences T cell migration to the lung, we examined fucosyltransferase-IV and –VII double knockout (FtDKO) mice. We discovered that FtDKO mice showed significant increases (∼5-fold) in numbers of naïve T cells in non-inflamed lung parenchyma with no evidence of induced bronchus-associated lymphoid tissue. In contrast, activated T cells were reduced in inflamed lungs of FtDKO mice following viral infection, consistent with the established role of selectin mediated T cell extravasation into inflamed lung. Adoptive transfer of T cells into FtDKO mice revealed impaired T cell entry to lymph nodes, but selective accumulation in non-lymphoid organs. Moreover, inhibition of T cell entry to the lymph nodes by blockade of L-selectin, or treatment of T cells with pertussis toxin to inhibit chemokine dependent G-coupled receptor signaling, also resulted in increased T cells in non-lymphoid organs. Conversely, inhibition of T cell egress from lymph nodes using FTY720 agonism of S1P1 impaired T cell migration into non-lymphoid organs. Conclusions/Significance Taken together, our results suggest that impaired T cell entry into lymph nodes via high endothelial venules due to genetic deficiency of selectin ligands results in the selective re-distribution and accumulation of T cells in non-lymphoid organs, and correlates with their increased frequency in the blood. Re-distribution of T cells into organs could potentially play a role in the initiation of T cell mediated organ diseases.
Collapse
Affiliation(s)
- John R. Harp
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Thandi M. Onami
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW IgE-mediated allergy can be treated by subcutaneous allergen-specific immunotherapy (SCIT). However, the percentage of allergic patients undergoing SCIT is low, mainly due to the long duration of the therapy and the risk of severe systemic allergic reactions associated with the allergen administration. Typically, SCIT requires dozens of subcutaneous allergen injections that stretch over 3-5 years. Over the last decade, sublingual immunotherapy has been established as an alternative to SCIT, but treatment duration and dosing frequencies could not be reduced. Recently, immunotherapy by direct administration of the allergen into lymph nodes [intralymphatic immunotherapy (ILIT)] has proven a promising alternative and this method is the focus of the present review. RECENT FINDINGS Several studies on animals and on humans have shown that direct injection into lymph nodes enhanced immune responses to protein, peptide, and naked DNA vaccines. Moreover, ILIT strongly improved allergen immunotherapy, so that the number of allergen administrations as well as the allergen dose could be reduced. As ILIT was also well tolerated, practically painless, and easy to perform, patient compliance was improved as compared with SCIT. SUMMARY Direct ILIT into a subcutaneous lymph node markedly enhances protective immune responses, so that both the dose and the number of allergen injections can be reduced, making ILIT safer and faster than other forms of immunotherapy, and most importantly, this enhances patient convenience and compliance.
Collapse
|
28
|
Hofmann J, Greter M, Du Pasquier L, Becher B. B-cells need a proper house, whereas T-cells are happy in a cave: the dependence of lymphocytes on secondary lymphoid tissues during evolution. Trends Immunol 2010; 31:144-53. [PMID: 20181529 DOI: 10.1016/j.it.2010.01.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Revised: 01/11/2010] [Accepted: 01/14/2010] [Indexed: 12/24/2022]
Abstract
A fundamental tenet of immunology is that adaptive immune responses are initiated in secondary lymphoid tissues. This dogma has been challenged by several recent reports. We discuss how successful T cell-mediated immunity can be initiated outside of such dedicated structures, whereas they are required for adaptive humoral immunity. This resembles an ancient immune pathway in the oldest cold-blooded vertebrates, which lack lymph nodes and sophisticated B-cell responses including optimal affinity maturation. The T-cell, however, has retained the capacity to recognize antigen in a lymph node-free environment. Besides bone marrow and lung, the liver is one organ that can potentially serve as a surrogate lymphoid organ and could represent a remnant from the time before lymph nodes developed.
Collapse
Affiliation(s)
- Janin Hofmann
- Division of Neuroimmunology, Inst. Exp. Immunology, Department of Pathology, University Hospital of Zurich, 8057 Zurich, Switzerland
| | | | | | | |
Collapse
|
29
|
|