1
|
Labeur-Iurman L, Harker JA. Mechanisms of antibody mediated immunity - Distinct in early life. Int J Biochem Cell Biol 2024; 172:106588. [PMID: 38768890 DOI: 10.1016/j.biocel.2024.106588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024]
Abstract
Immune responses in early life are characterized by a failure to robustly generate long-lasting protective responses against many common pathogens or upon vaccination. This is associated with a reduced ability to generate T-cell dependent high affinity antibodies. This review highlights the differences in T-cell dependent antibody responses observed between infants and adults, in particular focussing on the alterations in immune cell function that lead to reduced T follicular helper cell-B cell crosstalk within germinal centres in early life. Understanding the distinct functional characteristics of early life humoral immunity, and how these are regulated, will be critical in guiding age-appropriate immunological interventions in the very young.
Collapse
Affiliation(s)
- Lucia Labeur-Iurman
- National Heart & Lung Institute, Imperial College London, London, United Kingdom.
| | - James A Harker
- National Heart & Lung Institute, Imperial College London, London, United Kingdom; Centre for Paediatrics and Child Health, Imperial College London, London, United Kingdom.
| |
Collapse
|
2
|
Cully MD, Nolte JE, Patel A, Vaughan AE, May MJ. Loss of Lymphatic IKKα Disrupts Lung Immune Homeostasis, Drives BALT Formation, and Protects against Influenza. Immunohorizons 2024; 8:478-491. [PMID: 39007717 PMCID: PMC11294277 DOI: 10.4049/immunohorizons.2400047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 06/19/2024] [Indexed: 07/16/2024] Open
Abstract
IκB kinase (IKK)α controls noncanonical NF-κB signaling required for lymphoid organ development. We showed previously that lymph node formation is ablated in IkkαLyve-1 mice constitutively lacking IKKα in lymphatic endothelial cells (LECs). We now reveal that loss of IKKα in LECs leads to the formation of BALT in the lung. Tertiary lymphoid structures appear only in the lungs of IkkαLyve-1 mice and are not present in any other tissues, and these highly organized BALT structures form after birth and in the absence of inflammation. Additionally, we show that IkkαLyve-1 mice challenged with influenza A virus (IAV) exhibit markedly improved survival and reduced weight loss compared with littermate controls. Importantly, we determine that the improved morbidity and mortality of IkkαLyve-1 mice is independent of viral load and rate of clearance because both mice control and clear IAV infection similarly. Instead, we show that IFN-γ levels are decreased, and infiltration of CD8 T cells and monocytes into IkkαLyve-1 lungs is reduced. We conclude that ablating IKKα in LECs promotes BALT formation and reduces the susceptibility of IkkαLyve-1 mice to IAV infection through a decrease in proinflammatory stimuli.
Collapse
Affiliation(s)
- Michelle D. Cully
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Julianne E. Nolte
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Athena Patel
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Andrew E. Vaughan
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Michael J. May
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| |
Collapse
|
3
|
Si Y, Wang Y, Tian Q, Wang Q, Pollard JM, Srivastava PK, Esser-Kahn AP, Collier JH, Sperling AI, Chong AS. Lung cDC1 and cDC2 dendritic cells priming naive CD8 + T cells in situ prior to migration to draining lymph nodes. Cell Rep 2023; 42:113299. [PMID: 37864794 PMCID: PMC10676754 DOI: 10.1016/j.celrep.2023.113299] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/21/2023] [Accepted: 10/02/2023] [Indexed: 10/23/2023] Open
Abstract
The current paradigm indicates that naive T cells are primed in secondary lymphoid organs. Here, we present evidence that intranasal administration of peptide antigens appended to nanofibers primes naive CD8+ T cells in the lung independently and prior to priming in the draining mediastinal lymph node (MLN). Notably, comparable accumulation and transcriptomic responses of CD8+ T cells in lung and MLN are observed in both Batf3KO and wild-type (WT) mice, indicating that, while cDC1 dendritic cells (DCs) are the major subset for cross-presentation, cDC2 DCs alone are capable of cross-priming CD8+ T cells both in the lung and draining MLN. Transcription analyses reveal distinct transcriptional responses in lung cDC1 and cDC2 to intranasal nanofiber immunization. However, both DC subsets acquire shared transcriptional responses upon migration into the lymph node, thus uncovering a stepwise activation process of cDC1 and cDC2 toward their ability to cross-prime effector and functional memory CD8+ T cell responses.
Collapse
Affiliation(s)
- Youhui Si
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Department of Surgery, The University of Chicago, Chicago, IL 60637, USA.
| | - Yihan Wang
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Qiaomu Tian
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Qiang Wang
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Jared M Pollard
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Pramod K Srivastava
- Department of Immunology and Carole and Ray Neag Comprehensive Cancer Center, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Aaron P Esser-Kahn
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA
| | - Anne I Sperling
- Department of Medicine, Pulmonary and Critical Care, University of Virginia, Charlottesville, VA 22908, USA
| | - Anita S Chong
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
4
|
Parlak Ak T. Bronchus-Associated Lymphoid Tissue (BALT) Histology and Its Role in Various Pathologies. Vet Med Sci 2021. [DOI: 10.5772/intechopen.99366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The lower respiratory tract is in direct communication with the external environment for gas exchange to occur. Therefore, it is constantly exposed to allergens, antigens, bacteria, viruses, and a wide variety of airborne foreign bodies. Bronchus-associated lymphoid tissue (BALT), which develops in response to these exposures and is one of the most prominent representatives of mucosa-associated lymphoid tissue (MALT), is important for generating rapid and specific bronchopulmonary adaptive immune responses. Therefore, this chapter focuses on the lymphoid architecture of BALT, which was first discovered in the bronchial wall of rabbits, its inducible form called inducible BALT (iBALT), its immunological response mechanisms, and its roles in certain pathologies including infectious and autoimmune diseases as well as in allergic and malignant conditions. In conclusion, it is hypothesized that BALT plays an important role in maintaining health and in the development of lower respiratory tract diseases; thanks to the pulmonary immune system in which it functions as a functional lymphoid tissue.
Collapse
|
5
|
Jenkins MM, Bachus H, Botta D, Schultz MD, Rosenberg AF, León B, Ballesteros-Tato A. Lung dendritic cells migrate to the spleen to prime long-lived TCF1 hi memory CD8 + T cell precursors after influenza infection. Sci Immunol 2021; 6:eabg6895. [PMID: 34516781 DOI: 10.1126/sciimmunol.abg6895] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Meagan M Jenkins
- Division of Clinical Immunology and Rheumatology Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Holly Bachus
- Division of Clinical Immunology and Rheumatology Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Davide Botta
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael D Schultz
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alexander F Rosenberg
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.,Informatics Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Beatriz León
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - André Ballesteros-Tato
- Division of Clinical Immunology and Rheumatology Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
6
|
Nellore A, Killian JT, Porrett PM. Memory B Cells in Pregnancy Sensitization. Front Immunol 2021; 12:688987. [PMID: 34276679 PMCID: PMC8278195 DOI: 10.3389/fimmu.2021.688987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/10/2021] [Indexed: 11/13/2022] Open
Abstract
Memory B cells play an important role in immunity to pathogens as these cells are poised to rapidly differentiate into antibody-secreting cells upon antigen re-encounter. Memory B cells also develop over the course of HLA-sensitization during pregnancy and transplantation. In this review, we discuss the potential contribution of memory B cells to pregnancy sensitization as well as the impact of these cells on transplant candidacy and outcomes. We start by summarizing how B cell subsets are altered in pregnancy and discuss what is known about HLA-specific B cell responses given our current understanding of fetal antigen availability in maternal secondary lymphoid tissues. We then review the molecular mechanisms governing the generation and maintenance of memory B cells during infection - including the role of T follicular helper cells - and discuss the experimental evidence for the development of these cells during pregnancy. Finally, we discuss how memory B cells impact access to transplantation and transplant outcomes for a range of transplant recipients.
Collapse
Affiliation(s)
- Anoma Nellore
- Department of Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, AL, United States
| | - John T. Killian
- Department of Surgery, University of Alabama at Birmingham School of Medicine, Birmingham, AL, United States
| | - Paige M. Porrett
- Department of Surgery, University of Alabama at Birmingham School of Medicine, Birmingham, AL, United States
| |
Collapse
|
7
|
Friedrich SK, Schmitz R, Bergerhausen M, Lang J, Duhan V, Hardt C, Tenbusch M, Prinz M, Asano K, Bhat H, Hamdan TA, Lang PA, Lang KS. Replication of Influenza A Virus in Secondary Lymphatic Tissue Contributes to Innate Immune Activation. Pathogens 2021; 10:pathogens10050622. [PMID: 34069514 PMCID: PMC8160763 DOI: 10.3390/pathogens10050622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 11/23/2022] Open
Abstract
The replication of viruses in secondary lymphoid organs guarantees sufficient amounts of pattern-recognition receptor ligands and antigens to activate the innate and adaptive immune system. Viruses with broad cell tropism usually replicate in lymphoid organs; however, whether a virus with a narrow tropism relies on replication in the secondary lymphoid organs to activate the immune system remains not well studied. In this study, we used the artificial intravenous route of infection to determine whether Influenza A virus (IAV) replication can occur in secondary lymphatic organs (SLO) and whether such replication correlates with innate immune activation. Indeed, we found that IAV replicates in secondary lymphatic tissue. IAV replication was dependent on the expression of Sialic acid residues in antigen-presenting cells and on the expression of the interferon-inhibitor UBP43 (Usp18). The replication of IAV correlated with innate immune activation, resulting in IAV eradication. The genetic deletion of Usp18 curbed IAV replication and limited innate immune activation. In conclusion, we found that IAV replicates in SLO, a mechanism which allows innate immune activation.
Collapse
Affiliation(s)
- Sarah-Kim Friedrich
- Institute of Immunology, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany; (S.-K.F.); (R.S.); (M.B.); (J.L.); (V.D.); (C.H.); (H.B.)
| | - Rosa Schmitz
- Institute of Immunology, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany; (S.-K.F.); (R.S.); (M.B.); (J.L.); (V.D.); (C.H.); (H.B.)
| | - Michael Bergerhausen
- Institute of Immunology, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany; (S.-K.F.); (R.S.); (M.B.); (J.L.); (V.D.); (C.H.); (H.B.)
| | - Judith Lang
- Institute of Immunology, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany; (S.-K.F.); (R.S.); (M.B.); (J.L.); (V.D.); (C.H.); (H.B.)
| | - Vikas Duhan
- Institute of Immunology, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany; (S.-K.F.); (R.S.); (M.B.); (J.L.); (V.D.); (C.H.); (H.B.)
| | - Cornelia Hardt
- Institute of Immunology, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany; (S.-K.F.); (R.S.); (M.B.); (J.L.); (V.D.); (C.H.); (H.B.)
| | - Matthias Tenbusch
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany;
- Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, 79106 Freiburg, Germany
- Centre for NeuroModulation (NeuroModBasics), University of Freiburg, 79106 Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79106 Freiburg, Germany
| | - Kenichi Asano
- Laboratory of Immune Regulation, School of Life Science, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan;
| | - Hilal Bhat
- Institute of Immunology, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany; (S.-K.F.); (R.S.); (M.B.); (J.L.); (V.D.); (C.H.); (H.B.)
- Center for Molecular Medicine Cologne (CMMC), University Hospital Cologne, Robert Koch-Strasse 21, 50931 Köln, Germany
| | - Thamer A. Hamdan
- Institute of Immunology, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany; (S.-K.F.); (R.S.); (M.B.); (J.L.); (V.D.); (C.H.); (H.B.)
- Department of Medical Laboratories, Faculty of Health Sciences, American University of Madaba, Amman 11821, Jordan
- Correspondence: (T.A.H.); (K.S.L.)
| | - Philipp Alexander Lang
- Institute of Molecular Medicine II, University of Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany;
| | - Karl Sebastian Lang
- Institute of Immunology, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany; (S.-K.F.); (R.S.); (M.B.); (J.L.); (V.D.); (C.H.); (H.B.)
- Correspondence: (T.A.H.); (K.S.L.)
| |
Collapse
|
8
|
Truman LA, Bentley KL, Ruddle NH. Lymphotoxin targeted to salivary and lacrimal glands induces tertiary lymphoid organs and cervical lymphadenopathy and reduces tear production. Eur J Immunol 2020; 50:418-425. [PMID: 32012252 DOI: 10.1002/eji.201948300] [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: 07/09/2019] [Revised: 10/30/2019] [Accepted: 01/30/2020] [Indexed: 11/11/2022]
Abstract
To investigate the role of lymphotoxin (LT) in Sjögren's syndrome (SS) and in mucosal associated lymphoid tissue (MALT)-lymphoma, we made transgenic mice (Amy1-LTαβ) that targeted LTα and LTβ to the salivary and lacrimal glands. Amy1-LTαβ mice developed atrophic salivary and lacrimal glands that contained tertiary lymphoid organs (TLOs) and had reduced tear production. Amy1-LTαβ mice developed cervical lymphadenopathy but not MALT-lymphoma. TLO formation in the salivary and lacrimal glands of Amy1-LTαβ was not sufficient to induce autoimmunity as measured by autoantibody titres.
Collapse
Affiliation(s)
- Lucy A Truman
- ENT Department, West Suffolk Hospital, Hardwick Lane, Bury St Edmunds, UK.,Yale School of Public Health, New Haven, CT, USA
| | | | | |
Collapse
|
9
|
Wang S, Hirose S, Ghiasi H. The Absence of Lymphotoxin-α, a Herpesvirus Entry Mediator (HVEM) Ligand, Affects Herpes Simplex Virus 1 Infection In Vivo Differently than the Absence of Other HVEM Cellular Ligands. J Virol 2019; 93:e00707-19. [PMID: 31142672 PMCID: PMC6675894 DOI: 10.1128/jvi.00707-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 05/22/2019] [Indexed: 12/12/2022] Open
Abstract
Previously, we reported that the absence of herpesvirus entry mediator (HVEM) decreases latency but not primary infection in ocularly infected mice. Recently, we reported that similar to the absence of HVEM, the absence of HVEM ligands (i.e., LIGHT, CD160, and B and T lymphocyte attenuator [BTLA]) also decreased latency but not primary infection. Similar to LIGHT, CD160, and BTLA, another member of tumor necrosis factor (TNF) superfamily, lymphotoxin-α (LTα), also interacts with HVEM. To determine whether LTα decreases latency in infected mice, we ocularly infected LTα-/- mice with latency-associated transcript-positive [LAT(+)] and LAT(-) viruses using similarly infected wild-type (WT) mice as controls. In contrast to WT C57BL/6 mice, LTα-/- mice were highly susceptible to ocular herpes simplex virus 1 (HSV-1) infection, independent of the presence or absence of LAT. Survival was partially restored by adoptive transfer of CD4+, CD8+, or total T cells. Infected LTα-/- mice had significantly higher corneal scarring than WT mice, and adoptive T cell transfer did not alter the severity of eye disease. In contrast to results in WT mice, the amount of latency was not affected by the absence of LAT. The amount of LAT RNA in LTα-/- mice infected with LAT(+) virus was similar to that in WT mice, and adoptive T cell transfer did not alter LAT RNA levels in LTα-/- infected mice. Increased latency in the absence of LTα correlated with upregulation of HVEM, LIGHT, CD160, and BTLA transcripts as well as with an increase in markers of T cell exhaustion. The results of our study suggest that LTα has antipathogenic and anti-inflammatory functions and may act to protect the host from infection.IMPORTANCE Recently, we evaluated the effects of HVEM and its ligands (LIGHT, CD160, and BTLA) on HSV-1 infectivity. However, the effect of LTα, another member of the TNF superfamily, on HSV-1 latency and eye disease is not known. Here, we demonstrate increased latency and corneal scarring in LTα-/- infected mice, independent of the presence of LAT. In addition, infected mice were highly susceptible to HSV-1 infection, and survival was partially but not significantly restored by adoptive T cell transfer. These results suggest that the absence of LTα affects HSV-1 infectivity differently than the absence of HVEM, LIGHT, CD160, and BTLA.
Collapse
Affiliation(s)
- Shaohui Wang
- Center for Neurobiology and Vaccine Development, Department of Surgery, Ophthalmology Research, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Satoshi Hirose
- Center for Neurobiology and Vaccine Development, Department of Surgery, Ophthalmology Research, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Homayon Ghiasi
- Center for Neurobiology and Vaccine Development, Department of Surgery, Ophthalmology Research, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
10
|
Miyauchi K. Helper T Cell Responses to Respiratory Viruses in the Lung: Development, Virus Suppression, and Pathogenesis. Viral Immunol 2017. [DOI: 10.1089/vim.2017.0018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Kosuke Miyauchi
- RIKEN Center for Integrative Medical Science, Yokohama, Japan
| |
Collapse
|
11
|
Takahashi Y, Onodera T, Adachi Y, Ato M. Adaptive B Cell Responses to Influenza Virus Infection in the Lung. Viral Immunol 2017; 30:431-437. [PMID: 28661720 DOI: 10.1089/vim.2017.0025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Adaptive B cell response is a key arm of protective immunity against influenza viruses. Owing to the acutely infectious and cytopathic nature of these viruses, efficient containment of viral spread relies on the prompt provision of protective antibodies to the site of virus infection, the respiratory tract (RT). To accelerate the protective antibody response, B cell responses can be ectopically induced, maintained, and reactivated in the lungs after primary and secondary infection, thereby providing an anatomical advantage in supplying neutralizing antibodies against reinfecting viruses with faster kinetics. However, the prompt supply of protective antibodies may be insufficient to protect against reinfection because influenza viruses can easily change their antigenic profiles to escape antibody surveillance. B cell responses have multiple strategies for adjusting antibody repertoires according to viral fitness, one of which is the formation of local germinal centers capable of selecting B cell repertoires for antigenically subdominant, but conserved, epitopes. In this review, we discuss several unique aspects of B cell responses that take place at local sites to combat acutely infectious and rapidly mutating influenza viruses.
Collapse
Affiliation(s)
- Yoshimasa Takahashi
- Department of Immunology, National Institute of Infectious Diseases , Tokyo, Japan
| | - Taishi Onodera
- Department of Immunology, National Institute of Infectious Diseases , Tokyo, Japan
| | - Yu Adachi
- Department of Immunology, National Institute of Infectious Diseases , Tokyo, Japan
| | - Manabu Ato
- Department of Immunology, National Institute of Infectious Diseases , Tokyo, Japan
| |
Collapse
|
12
|
Koroleva EP, Fu YX, Tumanov AV. Lymphotoxin in physiology of lymphoid tissues - Implication for antiviral defense. Cytokine 2016; 101:39-47. [PMID: 27623349 DOI: 10.1016/j.cyto.2016.08.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/17/2016] [Accepted: 08/19/2016] [Indexed: 12/13/2022]
Abstract
Lymphotoxin (LT) is a member of the tumor necrosis factor (TNF) superfamily of cytokines which serves multiple functions, including the control of lymphoid organ development and maintenance, as well as regulation of inflammation and autoimmunity. Although the role of LT in organogenesis and maintenance of lymphoid organs is well established, the contribution of LT pathway to homeostasis of lymphoid organs during the immune response to pathogens is less understood. In this review, we highlight recent advances on the role of LT pathway in antiviral immune responses. We discuss the role of LT signaling in lymphoid organ integrity, type I IFN production and regulation of protection and immunopathology during viral infections. We further discuss the potential of therapeutic targeting LT pathway for controlling immunopathology and antiviral protection.
Collapse
Affiliation(s)
- Ekaterina P Koroleva
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas School of Medicine, UT Health Science Center, San Antonio, TX, USA; Trudeau Institute, Saranac Lake, NY
| | - Yang-Xin Fu
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alexei V Tumanov
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas School of Medicine, UT Health Science Center, San Antonio, TX, USA; Trudeau Institute, Saranac Lake, NY.
| |
Collapse
|
13
|
Hwang JY, Randall TD, Silva-Sanchez A. Inducible Bronchus-Associated Lymphoid Tissue: Taming Inflammation in the Lung. Front Immunol 2016; 7:258. [PMID: 27446088 PMCID: PMC4928648 DOI: 10.3389/fimmu.2016.00258] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/17/2016] [Indexed: 01/09/2023] Open
Abstract
Following pulmonary inflammation, leukocytes that infiltrate the lung often assemble into structures known as inducible Bronchus-Associated Lymphoid Tissue (iBALT). Like conventional lymphoid organs, areas of iBALT have segregated B and T cell areas, specialized stromal cells, high endothelial venules, and lymphatic vessels. After inflammation is resolved, iBALT is maintained for months, independently of inflammation. Once iBALT is formed, it participates in immune responses to pulmonary antigens, including those that are unrelated to the iBALT-initiating antigen, and often alters the clinical course of disease. However, the mechanisms that govern immune responses in iBALT and determine how iBALT impacts local and systemic immunity are poorly understood. Here, we review our current understanding of iBALT formation and discuss how iBALT participates in pulmonary immunity.
Collapse
Affiliation(s)
- Ji Young Hwang
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham , Birmingham, AL , USA
| | - Troy D Randall
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham , Birmingham, AL , USA
| | - Aaron Silva-Sanchez
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham , Birmingham, AL , USA
| |
Collapse
|
14
|
Sun T, Rojas OL, Li C, Philpott DJ, Gommerman JL. Hematopoietic LTβR deficiency results in skewed T cell cytokine profiles during a mucosal viral infection. J Leukoc Biol 2015; 100:103-10. [DOI: 10.1189/jlb.4mab0715-294r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 11/12/2015] [Indexed: 12/31/2022] Open
|
15
|
Wang Y, Sui Y, Kato S, Hogg AE, Steel JC, Morris JC, Berzofsky JA. Vaginal type-II mucosa is an inductive site for primary CD8⁺ T-cell mucosal immunity. Nat Commun 2015; 6:6100. [PMID: 25600442 PMCID: PMC4348041 DOI: 10.1038/ncomms7100] [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: 09/15/2014] [Accepted: 12/15/2014] [Indexed: 02/07/2023] Open
Abstract
The structured lymphoid tissues are considered the only inductive sites where primary T-cell immune responses occur. The naïve T cells in structured lymphoid tissues, once being primed by antigen-bearing dendritic cells, differentiate into memory T cells and traffic back to the mucosal sites through the bloodstream. Contrary to this belief, here we show that the vaginal type-II mucosa itself, despite the lack of structured lymphoid tissues, can act as an inductive site during primary CD8(+) T-cell immune responses. We provide evidence that the vaginal mucosa supports both the local immune priming of naïve CD8(+) T cells and the local expansion of antigen-specific CD8(+) T cells, thereby demonstrating a different paradigm for primary mucosal T-cell immune induction.
Collapse
Affiliation(s)
- Yichuan Wang
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland 20892, USA
| | - Yongjun Sui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland 20892, USA
| | - Shingo Kato
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland 20892, USA
| | - Alison E Hogg
- 1] Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland 20892, USA [2] Aeras, 1405 Research Boulevard, Rockville, Maryland 20850, USA
| | - Jason C Steel
- 1] The University of Queensland, Brisbane, Queensland 4120, Australia [2] Gallipoli Medical Research Foundation, Greenslopes, Queensland 4120, Australia
| | - John C Morris
- University of Cincinnati Cancer Institute, Cincinnati, Ohio 45267, USA
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
16
|
Whole inactivated avian Influenza H9N2 viruses induce nasal submucosal dendritic cells to sample luminal viruses via transepithelial dendrites and trigger subsequent DC maturation. Vaccine 2015; 33:1382-92. [PMID: 25613720 DOI: 10.1016/j.vaccine.2015.01.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 12/23/2014] [Accepted: 01/08/2015] [Indexed: 12/13/2022]
Abstract
Nasal mucosal barrier is a key impediment for the absorption of influenza whole inactivated virus (WIV) intranasal vaccine. Yet it is still unclear how WIV cross the epithelial cells (ECs) in nasal cavity. Here, in vitro, a coculture system was well established, consisting of surrogate nasal ECs (Calu-3) and dendritic cells (DCs). After adding H9N2 WIV on the apical side of ECs, we found that submucosal DCs extended their transepithelial dendrites (TEDs) and sampled luminal viruses. However, ECs were not involved in the transepithelial transport of viruses. Subsequently, the phenotypic and functional maturation of DCs were also enhanced, whereas they were attenuated after blocking of TED formation by anti-JAM1 antibody. In vivo, we confirmed that H9N2 WIV were capable of inducing nasal submucosal DCs to sample luminal viruses via TEDs in the nasal passage but not nasal-associated lymphoid tissue (NALT). CD103(+) and CD103(-) DC subsets participated in this process. Of note, chemokine CCL20, released from the H9N2 WIV-induced ECs, played a vital role in DC recruitment and TED formation. Taken together, our findings indicated that TEDs played a critical role in facilitating viral transport across the epithelial barrier, which may guide the design of novel nasal mucosal vaccine strategies.
Collapse
|
17
|
Lucchesi D, Pitzalis C, Bombardieri M. EBV and other viruses as triggers of tertiary lymphoid structures in primary Sjögren's syndrome. Expert Rev Clin Immunol 2014; 10:445-55. [PMID: 24564506 DOI: 10.1586/1744666x.2014.892417] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Sjögren's syndrome (SS) is an autoimmune disease that targets salivary (SG) and lachrymal glands, leading to exocrine dysfunction. Several viruses have been associated with SS, although the role of persistent viral infections in triggering and/or perpetuating the disease is still a matter of controversy. Together with exocrine dysfunction, SS is characterised by the production of autoantibodies and the presence of lymphomonocytic periductal aggregates in the SG, which in 30/40% of the patients display features of tertiary lymphoid structures (TLS) supporting an ectopic germinal centre response. Here we first review i) the relevance of TLS in SS and ii) the evidence in support of a role for viruses in SS insurgence and/or persistence; next, iii) we review recent data which links viral infection with TLS formation in the SG and suggests that viral-host interactions within TLS favour breach of tolerance and development of autoimmunity in SS.
Collapse
Affiliation(s)
- Davide Lucchesi
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | | | | |
Collapse
|
18
|
Macdonald DC, Singh H, Whelan MA, Escors D, Arce F, Bottoms SE, Barclay WS, Maini M, Collins MK, Rosenberg WMC. Harnessing alveolar macrophages for sustained mucosal T-cell recall confers long-term protection to mice against lethal influenza challenge without clinical disease. Mucosal Immunol 2014; 7:89-100. [PMID: 23715172 DOI: 10.1038/mi.2013.27] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 04/01/2013] [Indexed: 02/04/2023]
Abstract
Vaccines that induce T cells, which recognize conserved viral proteins, could confer universal protection against seasonal and pandemic influenza strains. An effective vaccine should generate sufficient mucosal T cells to ensure rapid viral control before clinical disease. However, T cells may also cause lung injury in influenza, so this approach carries inherent risks. Here we describe intranasal immunization of mice with a lentiviral vector expressing influenza nucleoprotein (NP), together with an NFκB activator, which transduces over 75% of alveolar macrophages (AM). This strategy recalls and expands NP-specific CD8+ T cells in the lung and airway of mice that have been immunized subcutaneously, or previously exposed to influenza. Granzyme B-high, lung-resident T-cell populations persist for at least 4 months and can control a lethal influenza challenge without harmful cytokine responses, weight loss, or lung injury. These data demonstrate that AM can be harnessed as effective antigen-presenting cells for influenza vaccination.
Collapse
Affiliation(s)
- D C Macdonald
- Division of Infection and Immunity and MRC Centre for Medical Molecular Virology, University College London, London, UK
| | - H Singh
- Division of Infection and Immunity and MRC Centre for Medical Molecular Virology, University College London, London, UK
| | - M A Whelan
- Division of Medicine, University College London, London, UK
| | - D Escors
- Division of Infection and Immunity and MRC Centre for Medical Molecular Virology, University College London, London, UK
| | - F Arce
- Division of Infection and Immunity and MRC Centre for Medical Molecular Virology, University College London, London, UK
| | - S E Bottoms
- Division of Medicine, University College London, London, UK
| | - W S Barclay
- Division of Virology, Imperial College London, St Mary's Campus, London, UK
| | - M Maini
- Division of Infection and Immunity and MRC Centre for Medical Molecular Virology, University College London, London, UK
| | - M K Collins
- Division of Infection and Immunity and MRC Centre for Medical Molecular Virology, University College London, London, UK
| | | |
Collapse
|
19
|
Kato A, Hulse KE, Tan BK, Schleimer RP. B-lymphocyte lineage cells and the respiratory system. J Allergy Clin Immunol 2013; 131:933-57; quiz 958. [PMID: 23540615 DOI: 10.1016/j.jaci.2013.02.023] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 02/20/2013] [Accepted: 02/21/2013] [Indexed: 12/12/2022]
Abstract
Adaptive humoral immune responses in the airways are mediated by B cells and plasma cells that express highly evolved and specific receptors and produce immunoglobulins of most isotypes. In some cases, such as autoimmune diseases or inflammatory diseases caused by excessive exposure to foreign antigens, these same immune cells can cause disease by virtue of overly vigorous responses. This review discusses the generation, differentiation, signaling, activation, and recruitment pathways of B cells and plasma cells, with special emphasis on unique characteristics of subsets of these cells functioning within the respiratory system. The primary sensitization events that generate B cells responsible for effector responses throughout the airways usually occur in the upper airways, tonsils, and adenoid structures that make up the Waldeyer ring. On secondary exposure to antigen in the airways, antigen-processing dendritic cells migrate into secondary lymphoid organs, such as lymph nodes, that drain the upper and lower airways, and further B-cell expansion takes place at those sites. Antigen exposure in the upper or lower airways can also drive expansion of B-lineage cells in the airway mucosal tissue and lead to the formation of inducible lymphoid follicles or aggregates that can mediate local immunity or disease.
Collapse
Affiliation(s)
- Atsushi Kato
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | |
Collapse
|
20
|
Abstract
Evidence has increasingly shown that the lungs are a major site of immune regulation. A robust and highly regulated immune response in the lung protects the host from pathogen infection, whereas an inefficient or deleterious response can lead to various pulmonary diseases. Many cell types, such as epithelial cells, dendritic cells, macrophages, neutrophils, eosinophils, and B and T lymphocytes, contribute to lung immunity. This review focuses on the recent advances in understanding how T lymphocytes mediate pulmonary host defenses against bacterial, viral, and fungal pathogens.
Collapse
Affiliation(s)
- Kong Chen
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15201, USA
| | | |
Collapse
|
21
|
O’Meara CP, Armitage CW, Harvie MCG, Timms P, Lycke NY, Beagley KW. Immunization with a MOMP-based vaccine protects mice against a pulmonary Chlamydia challenge and identifies a disconnection between infection and pathology. PLoS One 2013; 8:e61962. [PMID: 23613984 PMCID: PMC3628704 DOI: 10.1371/journal.pone.0061962] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 03/16/2013] [Indexed: 12/31/2022] Open
Abstract
Chlamydia pneumoniae is responsible for up to 20% of community acquired pneumonia and can exacerbate chronic inflammatory diseases. As the majority of infections are either mild or asymptomatic, a vaccine is recognized to have the greatest potential to reduce infection and disease prevalence. Using the C. muridarum mouse model of infection, we immunized animals via the intranasal (IN), sublingual (SL) or transcutaneous (TC) routes, with recombinant chlamydial major outer membrane protein (MOMP) combined with adjuvants CTA1-DD or a combination of cholera toxin/CpG-oligodeoxynucleotide (CT/CpG). Vaccinated animals were challenged IN with C. muridarum and protection against infection and pathology was assessed. SL and TC immunization with MOMP and CT/CpG was the most protective, significantly reducing chlamydial burden in the lungs and preventing weight loss, which was similar to the protection induced by a previous live infection. Unlike a previous infection however, these vaccinations also provided almost complete protection against fibrotic scarring in the lungs. Protection against infection was associated with antigen-specific production of IFNγ, TNFα and IL-17 by splenocytes, however, protection against both infection and pathology required the induction of a similar pro-inflammatory response in the respiratory tract draining lymph nodes. Interestingly, we also identified two contrasting vaccinations capable of preventing infection or pathology individually. Animals IN immunized with MOMP and either adjuvant were protected from infection, but not the pathology. Conversely, animals TC immunized with MOMP and CTA1-DD were protected from pathology, even though the chlamydial burden in this group was equivalent to the unimmunized controls. This suggests that the development of pathology following an IN infection of vaccinated animals was independent of bacterial load and may have been driven instead by the adaptive immune response generated following immunization. This identifies a disconnection between the control of infection and the development of pathology, which may influence the design of future vaccines.
Collapse
Affiliation(s)
- Connor P. O’Meara
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - Charles W. Armitage
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - Marina C. G. Harvie
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - Peter Timms
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - Nils Y. Lycke
- Mucosal Immunobiology and Vaccine Centre (MIVAC), University of Göteborg, Göteborg, Götaland, Sweden
| | - Kenneth W. Beagley
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
- * E-mail:
| |
Collapse
|
22
|
Delayed but effective induction of mucosal memory immune responses against genital HSV-2 in the absence of secondary lymphoid organs. Mucosal Immunol 2013; 6:56-68. [PMID: 22718264 DOI: 10.1038/mi.2012.48] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
To examine whether local immunization in the absence of secondary lymphoid organs (SLOs) could establish effective antiviral memory responses in the female genital tract, we examined immunity in the vaginal tracts of LTα-/- mice, LTα-/- SPL (splenectomized), and control C57BL/6 (WT) mice. All three groups of mice were immunized intravaginally (IVAG) with attenuated thymidine kinase-negative (TK(-)) Herpes simplex virus type 2 (HSV-2) and challenged 4-6 weeks later with wild-type (WT) HSV-2. Both groups of LTα-/- mice exhibited delayed viral clearance and prolonged genital pathology after immunization. Following IVAG WT HSV-2 challenge, LTα-/- and LTα-/- SPL mice had significantly lower levels of HSV-2-specific IgG and IgA in the vaginal secretions. Although the frequency of B and T cells in the vaginal mucosa was comparable or higher in both groups of LTα-/- mice, lower frequency of HSV-2-specific interferon-γ (IFNγ)-producing CD3+ T cells was seen after immunization and after challenge, compared with WT group. Despite this, immunized mice in all three groups showed complete sterile protection against IVAG WT HSV-2 challenge. These results show that even in the absence of SLOs, IVAG immunization generates effector memory immune responses at genital mucosa that can provide antiviral protection against subsequent viral exposures. This will inform new strategies to design mucosal vaccines against sexually transmitted infections.
Collapse
|
23
|
Boyden AW, Legge KL, Waldschmidt TJ. Pulmonary infection with influenza A virus induces site-specific germinal center and T follicular helper cell responses. PLoS One 2012; 7:e40733. [PMID: 22792401 PMCID: PMC3394713 DOI: 10.1371/journal.pone.0040733] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 06/12/2012] [Indexed: 11/19/2022] Open
Abstract
Protection from influenza A virus (IAV) challenge requires switched, high affinity Abs derived from long-lived memory B cells and plasma cells. These B cell subsets are generated in germinal centers (GCs), hallmark structures of T helper cell-driven B cell immunity. A full understanding of the GC reaction after respiratory IAV infection is lacking, as is the characterization of T follicular helper (TFH) cells that support GCs. Here, GC B cell and TFH cell responses were studied in mice following pulmonary challenge with IAV. Marked GC reactions were induced in draining lymph nodes (dLNs), lung, spleen and nasal-associated lymphoid tissue (NALT), although the magnitude and kinetics of the response was site-specific. Examination of switching within GCs demonstrated IgG2+ cells to compose the largest fraction in dLNs, lung and spleen. IgA+ GC B cells were infrequent in these sites, but composed a significant subset of the switched GC population in NALT. Further experiments demonstrated splenectomized mice to withstand a lethal recall challenge, suggesting the spleen to be unnecessary for long-term protection in spite of strong GC responses in this organ. Final studies showed that TFH cell numbers were highest in dLNs and spleen, and peaked in all sites prior to the height of the GC reaction. TFH cells purified from dLNs generated IL-21 and IFNγ upon activation, although CD4+CXCR5− T effector cells produced higher levels of all cytokines. Collectively, these findings reveal respiratory IAV infection to induce strong T helper cell-driven B cell responses in various organs, with each site displaying unique attributes.
Collapse
Affiliation(s)
- Alexander W. Boyden
- Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
| | - Kevin L. Legge
- Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
- Department of Microbiology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
| | - Thomas J. Waldschmidt
- Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
24
|
De Trez C. Lymphotoxin-beta receptor expression and its related signaling pathways govern dendritic cell homeostasis and function. Immunobiology 2012; 217:1250-8. [PMID: 22795648 DOI: 10.1016/j.imbio.2012.06.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 06/20/2012] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DCs) play a fundamental function, either positive or detrimental, in regulating immune responses. Numerous specialized DC subsets exist in different organs. However, the trophic factors regulating their origin, location, homeostasis and functions remains to be fully understood. Recent evidence indicates that signaling via the lymphotoxin β receptor (LTβR) can function as a trophic signaling system for specific DCs. LTβR is part of a complex signaling network that provides both positive and inhibitory signals to DC subsets. In this review, we focus on the role of LTβR expressed in DC subsets and its associated signaling pathways that regulate DC homeostasis and function. Therapeutically targeting the LTβR signaling pathway could support the development of a beneficial immune response for the host.
Collapse
Affiliation(s)
- Carl De Trez
- VIB Department of Structural Biology, Laboratory of Cellular and Molecular Immunology, Vrij Universiteit Brussel (VUB), Building E8.01, Pleinlaan 2, B-1050 Brussels, Belgium.
| |
Collapse
|
25
|
Functional identification of dendritic cells in the teleost model, rainbow trout (Oncorhynchus mykiss). PLoS One 2012; 7:e33196. [PMID: 22427987 PMCID: PMC3299753 DOI: 10.1371/journal.pone.0033196] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 02/06/2012] [Indexed: 01/08/2023] Open
Abstract
Dendritic cells are specialized antigen presenting cells that bridge innate and adaptive immunity in mammals. This link between the ancient innate immune system and the more evolutionarily recent adaptive immune system is of particular interest in fish, the oldest vertebrates to have both innate and adaptive immunity. It is unknown whether dendritic cells co-evolved with the adaptive response, or if the connection between innate and adaptive immunity relied on a fundamentally different cell type early in evolution. We approached this question using the teleost model organism, rainbow trout (Oncorhynchus mykiss), with the aim of identifying dendritic cells based on their ability to stimulate naïve T cells. Adapting mammalian protocols for the generation of dendritic cells, we established a method of culturing highly motile, non-adherent cells from trout hematopoietic tissue that had irregular membrane processes and expressed surface MHCII. When side-by-side mixed leukocyte reactions were performed, these cells stimulated greater proliferation than B cells or macrophages, demonstrating their specialized ability to present antigen and therefore their functional homology to mammalian dendritic cells. Trout dendritic cells were then further analyzed to determine if they exhibited other features of mammalian dendritic cells. Trout dendritic cells were found to have many of the hallmarks of mammalian DCs including tree-like morphology, the expression of dendritic cell markers, the ability to phagocytose small particles, activation by toll-like receptor-ligands, and the ability to migrate in vivo. As in mammals, trout dendritic cells could be isolated directly from the spleen, or larger numbers could be derived from hematopoietic tissue and peripheral blood mononuclear cells in vitro.
Collapse
|
26
|
Gommerman JL, Summers deLuca L. LTβR and CD40: working together in dendritic cells to optimize immune responses. Immunol Rev 2012; 244:85-98. [PMID: 22017433 DOI: 10.1111/j.1600-065x.2011.01056.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Generating an immune response tailored to destroy an infecting organism while limiting bystander damage involves guiding T-cell activation using a variety of cues taken from the immunogen (antigen type, dose, and persistence, accompanying danger signals) as well as the host (tissue environment, T-cell frequency, and affinity for antigen). Dendritic cells (DCs) serve as translators of much of this information and are critically required for effective pathogen and tumor clearance. Moreover, dysregulation of DC activation can lead to autoimmunity. Inhibition of the lymphotoxin (LT) and CD40 pathways has been shown to be effective at quieting inflammation in settings where DC-T-cell interactions are key instigators of disease progression. In this review, we compare and contrast the CD40 and LT pathways in the context of receptor/ligand expression, signal transduction, and DC biology. We provide evidence that these two pathways play complementary roles in DC cytokine secretion, thus indirectly shaping the nature of the CD8(+) T-cell response to foreign antigen. Given the distinct role of these pathways in the context of DC function, we propose that dual therapies targeted at both the CD40 and LTβ receptor may have therapeutic potential in silencing DC-driven autoimmunity or in promoting tumor clearance.
Collapse
|
27
|
Wu H, Kumar A, Miao H, Holden-Wiltse J, Mosmann TR, Livingstone AM, Belz GT, Perelson AS, Zand MS, Topham DJ. Modeling of influenza-specific CD8+ T cells during the primary response indicates that the spleen is a major source of effectors. THE JOURNAL OF IMMUNOLOGY 2011; 187:4474-82. [PMID: 21948988 DOI: 10.4049/jimmunol.1101443] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The biological parameters that determine the distribution of virus-specific CD8(+) T cells during influenza infection are not all directly measurable by experimental techniques but can be inferred through mathematical modeling. Mechanistic and semimechanistic ordinary differential equations were developed to describe the expansion, trafficking, and disappearance of activated virus-specific CD8(+) T cells in lymph nodes, spleens, and lungs of mice during primary influenza A infection. An intensive sampling of virus-specific CD8(+) T cells from these three compartments was used to inform the models. Rigorous statistical fitting of the models to the experimental data allowed estimation of important biological parameters. Although the draining lymph node is the first tissue in which Ag-specific CD8(+) T cells are detected, it was found that the spleen contributes the greatest number of effector CD8(+) T cells to the lung, with rates of expansion and migration that exceeded those of the draining lymph node. In addition, models that were based on the number and kinetics of professional APCs fit the data better than those based on viral load, suggesting that the immune response is limited by Ag presentation rather than the amount of virus. Modeling also suggests that loss of effector T cells from the lung is significant and time dependent, increasing toward the end of the acute response. Together, these efforts provide a better understanding of the primary CD8(+) T cell response to influenza infection, changing the view that the spleen plays a minor role in the primary immune response.
Collapse
Affiliation(s)
- Hulin Wu
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Ding X, Yang W, Shi X, Du P, Su L, Qin Z, Chen J, Deng H. TNF receptor 1 mediates dendritic cell maturation and CD8 T cell response through two distinct mechanisms. THE JOURNAL OF IMMUNOLOGY 2011; 187:1184-91. [PMID: 21709152 DOI: 10.4049/jimmunol.1002902] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
TNF-α and its two receptors (TNFR1 and 2) are known to stimulate dendritic cell (DC) maturation and T cell response. However, the specific receptor and mechanisms involved in vivo are still controversial. In this study, we show that in response to an attenuated mouse hepatitis virus infection, DCs fail to mobilize and up-regulate CD40, CD80, CD86, and MHC class I in TNFR1(-/-) mice as compared with the wild-type and TNFR2(-/-) mice. Correspondingly, virus-specific CD8 T cell response was dramatically diminished in TNFR1(-/-) mice. Adoptive transfer of TNFR1-expressing DCs into TNFR1(-/-) mice rescues CD8 T cell response. Interestingly, adoptive transfer of TNFR1-expressing naive T cells also restores DC mobilization and maturation and endogenous CD8 T cell response. These results show that TNFR1, not TNFR2, mediates TNF-α stimulation of DC maturation and T cell response to mouse hepatitis virus in vivo. They also suggest two mechanisms by which TNFR1 mediates TNF-α-driven DC maturation, as follows: a direct effect through TNFR1 expressed on immature DCs and an indirect effect through TNFR1 expressed on naive T cells.
Collapse
Affiliation(s)
- Xilai Ding
- CAS Key Laboratory of Infection and Immunity, Center for Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | | | | | | | | | | | | | | |
Collapse
|
29
|
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: 20] [Impact Index Per Article: 1.4] [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
|
30
|
Kim DY, Sato A, Fukuyama S, Sagara H, Nagatake T, Kong IG, Goda K, Nochi T, Kunisawa J, Sato S, Yokota Y, Lee CH, Kiyono H. The airway antigen sampling system: respiratory M cells as an alternative gateway for inhaled antigens. THE JOURNAL OF IMMUNOLOGY 2011; 186:4253-62. [PMID: 21357262 DOI: 10.4049/jimmunol.0903794] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In this study, we demonstrated a new airway Ag sampling site by analyzing tissue sections of the murine nasal passages. We revealed the presence of respiratory M cells, which had the ability to take up OVA and recombinant Salmonella typhimurium expressing GFP, in the turbinates covered with single-layer epithelium. These M cells were also capable of taking up respiratory pathogen group A Streptococcus after nasal challenge. Inhibitor of DNA binding/differentiation 2 (Id2)-deficient mice, which are deficient in lymphoid tissues, including nasopharynx-associated lymphoid tissue, had a similar frequency of M cell clusters in their nasal epithelia to that of their littermates, Id2(+/-) mice. The titers of Ag-specific Abs were as high in Id2(-/-) mice as in Id2(+/-) mice after nasal immunization with recombinant Salmonella-ToxC or group A Streptococcus, indicating that respiratory M cells were capable of sampling inhaled bacterial Ag to initiate an Ag-specific immune response. Taken together, these findings suggest that respiratory M cells act as a nasopharynx-associated lymphoid tissue-independent alternative gateway for Ag sampling and subsequent induction of Ag-specific immune responses in the upper respiratory tract.
Collapse
Affiliation(s)
- Dong-Young Kim
- Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Bronchus-associated lymphoid tissue (BALT) is a constitutive mucosal lymphoid tissue adjacent to major airways in some mammalian species, including rats and rabbits, but not humans or mice. A related tissue, inducible BALT (iBALT), is an ectopic lymphoid tissue that is formed upon inflammation or infection in both mice and humans and can be found throughout the lung. Both BALT and iBALT acquire antigens from the airways and initiate local immune responses and maintain memory cells in the lungs. Here, we discuss the development and function of BALT and iBALT in the context of pulmonary immunity to infectious agents, tumors, and allergens as well as autoimmunity and inflammatory diseases of the lung.
Collapse
Affiliation(s)
- Troy D Randall
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
32
|
LTβR signaling in dendritic cells induces a type I IFN response that is required for optimal clonal expansion of CD8+ T cells. Proc Natl Acad Sci U S A 2011; 108:2046-51. [PMID: 21245292 DOI: 10.1073/pnas.1014188108] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
During an immune response, antigen-bearing dendritic cells (DCs) migrate to the local draining lymph node and present antigen to CD4(+) helper T cells. Antigen-activated CD4(+) T cells then up-regulate TNF superfamily members including CD40 ligand and lymphotoxin (LT)αβ. Although it is well-accepted that CD40 stimulation on DCs is required for DC licensing and cross-priming of CD8(+) T-cell responses, it is likely that other signals are integrated into a comprehensive DC activation program. Here we show that a cognate interaction between LTαβ on CD4(+) helper T cells and LTβ receptor on DCs results in unique signals that are necessary for optimal CD8(+) T-cell expansion via a type I IFN-dependent mechanism. In contrast, CD40 signaling appears to be more critical for CD8(+) T-cell IFNγ production. Therefore, different TNF family members provide integrative signals that shape the licensing potential of antigen-presenting DCs.
Collapse
|
33
|
Salek-Ardakani S, Croft M. Tumor necrosis factor receptor/tumor necrosis factor family members in antiviral CD8 T-cell immunity. J Interferon Cytokine Res 2010; 30:205-18. [PMID: 20377415 DOI: 10.1089/jir.2010.0026] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
CD8 memory T cells can play a critical role in protection against repeated exposure to infectious agents such as viruses, yet can also contribute to the immunopathology associated with these pathogens. Understanding the mechanisms that control effective memory responses has important ramifications for vaccine design and in the management of adverse immune reactions. Recent studies have implicated several members of the tumor necrosis factor receptor (TNFR) family as key stimulatory and inhibitory molecules involved in the regulation of CD8 T cells. In this review, we discuss their control of the generation, persistence, and reactivation of CD8 T cells during virus infection.
Collapse
Affiliation(s)
- Shahram Salek-Ardakani
- Division of Molecular Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA.
| | | |
Collapse
|
34
|
Kallal LE, Hartigan AJ, Hogaboam CM, Schaller MA, Lukacs NW. Inefficient lymph node sensitization during respiratory viral infection promotes IL-17-mediated lung pathology. THE JOURNAL OF IMMUNOLOGY 2010; 185:4137-47. [PMID: 20805422 DOI: 10.4049/jimmunol.1000677] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Development of bronchus-associated lymphoid tissue has been suggested to enhance local antiviral immune responses; however, ectopic lymph node formation often corresponds to chronic inflammatory diseases. These studies investigated the role of ectopic pulmonary lymph nodes upon respiratory syncytial virus (RSV) infection using CCR7-deficient mice, which develop bronchus-associated lymphoid tissue early in life. CCR7(-/-) mice exhibited impaired secondary lymph node formation, enhanced effector T cell responses and pathogenic mucus production in the lung after RSV infection. IL-17 production from CD4 T cells in CCR7(-/-) mice was most remarkably enhanced. Wild-type animals reconstituted with CCR7(-/-) bone marrow recapitulated the pathogenic lung phenotype in CCR7(-/-) mice, whereas CCR7(-/-) animals reconstituted with wild-type bone marrow had normal lymph node development, diminished IL-17 production and reduced lung pathology. Mixed bone marrow chimeras revealed an alteration of immune responses only in CCR7(-/-) T cells, suggesting that impaired trafficking promotes local effector cell generation. Lymphotoxin-α-deficient mice infected with RSV were used to further examine locally induced immune responses and demonstrated increased mucus production and amplified cytokine responses in the lung, especially IL-17. Neutralization of IL-17 in CCR7(-/-) or in lymphotoxin-α-deficient animals specifically inhibited mucus hypersecretion and reduced IL-13. Thus, immune cell trafficking to secondary lymph nodes is necessary for appropriate cytokine responses to RSV as well as modulation of the local environment.
Collapse
Affiliation(s)
- Lara E Kallal
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | |
Collapse
|
35
|
Day TA, Koch M, Nouailles G, Jacobsen M, Kosmiadi GA, Miekley D, Kuhlmann S, Jörg S, Gamradt P, Mollenkopf HJ, Hurwitz R, Reece ST, Kaufmann SHE, Kursar M. Secondary lymphoid organs are dispensable for the development of T-cell-mediated immunity during tuberculosis. Eur J Immunol 2010; 40:1663-73. [PMID: 20222088 DOI: 10.1002/eji.201040299] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Tuberculosis causes 2 million deaths per year, yet in most cases the immune response successfully contains the infection and prevents disease outbreak. Induced lymphoid structures associated with pulmonary granuloma are observed during tuberculosis in both humans and mice and could orchestrate host defense. To investigate whether granuloma perform lymphoid functions, mice lacking secondary lymphoid organs (SLO) were infected with Mycobacterium tuberculosis (MTB). As in WT mice, granuloma developed, exponential growth of MTB was controlled, and antigen-specific T-cell responses including memory T cells were generated in the absence of SLO. Moreover, adoptively transferred T cells were primed locally in lungs in a granuloma-dependent manner. T-cell activation was delayed in the absence of SLO, but resulted in a normal development program including protective subsets and functional recall responses that protected mice against secondary MTB infection. Our data demonstrate that protective immune responses can be generated independently of SLO during MTB infection and implicate local pulmonary T-cell priming as a mechanism contributing to host defense.
Collapse
Affiliation(s)
- Tracey A Day
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Yewdell JW. Designing CD8+ T cell vaccines: it's not rocket science (yet). Curr Opin Immunol 2010; 22:402-10. [PMID: 20447814 PMCID: PMC2908899 DOI: 10.1016/j.coi.2010.04.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Accepted: 04/12/2010] [Indexed: 01/09/2023]
Abstract
CD8+ T cells play important roles in clearing viral infections and eradicating tumors. Designing vaccines that elicit effective CD8+ T cell responses requires a thorough knowledge of the pathways of antigen presentation in vivo. Here, I review recent progress in understanding the activation of naïve CD8+ T cells in vivo, with particular emphasis on cross-priming, the presentation of protein antigens acquired by dendritic cells from their environment. With the rapid advances in this area of research, the dawn of rational vaccine design is at hand.
Collapse
|
37
|
Lipscomb MF, Hutt J, Lovchik J, Wu T, Lyons CR. The pathogenesis of acute pulmonary viral and bacterial infections: investigations in animal models. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2010; 5:223-52. [PMID: 19824827 DOI: 10.1146/annurev-pathol-121808-102153] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Acute viral and bacterial infections in the lower respiratory tract are major causes of morbidity and mortality worldwide. The proper study of pulmonary infections requires interdisciplinary collaboration among physicians and biomedical scientists to develop rational hypotheses based on clinical studies and to test these hypotheses in relevant animal models. Animal models for common lung infections are essential to understand pathogenic mechanisms and to clarify general mechanisms for host protection in pulmonary infections, as well as to develop vaccines and therapeutics. Animal models for uncommon pulmonary infections, such as those that can be caused by category A biothreat agents, are also very important because the infrequency of these infections in humans limits in-depth clinical studies. This review summarizes our understanding of innate and adaptive immune mechanisms in the lower respiratory tract and discusses how animal models for selected pulmonary pathogens can contribute to our understanding of the pathogenesis of lung infections and to the search for new vaccines and therapies.
Collapse
Affiliation(s)
- Mary F Lipscomb
- Departments of Pathology and University of New Mexico School of Medicine, Albuquerque, New Mexico 87131.
| | | | | | | | | |
Collapse
|
38
|
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: 3.2] [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
|
39
|
Prophylactic administration of bacterially derived immunomodulators improves the outcome of influenza virus infection in a murine model. J Virol 2010; 84:2983-95. [PMID: 20053748 DOI: 10.1128/jvi.01805-09] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Prophylactic or therapeutic immunomodulation is an antigen-independent strategy that induces nonspecific immune system activation, thereby enhancing host defense to disease. In this study, we investigated the effect of prophylactic immunomodulation on the outcome of influenza virus infection using three bacterially derived immune-enhancing agents known for promoting distinct immunological profiles. BALB/c mice were treated nasally with either cholera toxin (CT), a mutant form of the CT-related Escherichia coli heat-labile enterotoxin designated LT(R192G), or CpG oligodeoxynucleotide. Mice were subsequently challenged with a lethal dose of influenza A/PR/8/34 virus 24 h after the last immunomodulation treatment and either monitored for survival or sacrificed postchallenge for viral and immunological analysis. Treatment with the three immunomodulators prevented or delayed mortality and weight loss, but only CT and LT(R192G) significantly reduced initial lung viral loads as measured by plaque assay. Analysis performed 4 days postinfection indicated that prophylactic treatments with CT, LT(R192G), or CpG resulted in significantly increased numbers of CD4 T cells, B cells, and dendritic cells and altered costimulatory marker expression in the airways of infected mice, coinciding with reduced expression of pulmonary chemokines and the appearance of inducible bronchus-associated lymphoid tissue-like structures in the lungs. Collectively, these results suggest that, despite different immunomodulatory mechanisms, CT, LT(R192G), and CpG induce an initial inflammatory process and enhance the immune response to primary influenza virus challenge while preventing potentially damaging chemokine expression. These studies provide insight into the immunological parameters and immune modulation strategies that have the potential to enhance the nonspecific host response to influenza virus infection.
Collapse
|
40
|
Kocks JR, Adler H, Danzer H, Hoffmann K, Jonigk D, Lehmann U, Förster R. Chemokine receptor CCR7 contributes to a rapid and efficient clearance of lytic murine gamma-herpes virus 68 from the lung, whereas bronchus-associated lymphoid tissue harbors virus during latency. THE JOURNAL OF IMMUNOLOGY 2009; 182:6861-9. [PMID: 19454682 DOI: 10.4049/jimmunol.0801826] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Murine gamma-herpes virus 68 is a natural rodent pathogen closely related to the human gamma-herpes viruses Kaposi's sarcoma-associated herpes virus and EBV. By intranasally infecting wild-type and CCR7-deficient mice, we investigated whether CCR7 is necessary for viral clearance from the lung and the establishment of latency. We found during the lytic phase of infection that inflammation in lungs of CCR7(-/-) mice was more severe and viral load significantly higher compared with wild-type littermates. In addition, activation of T cells was delayed and clearance of the inflammation was retarded in mutant lungs, demonstrating that CCR7 is necessary for a rapid and efficient immune response. However, for the establishment of splenomegaly and latency, the presence of CCR7 was dispensable. Finally, by microdissecting BALT, we could demonstrate that these ectopic lymphoid structures are a place in the lung where virus resides during latency.
Collapse
Affiliation(s)
- Jessica R Kocks
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
SUMMARY Cytokines mediate key communication pathways essential for regulation of immune responses. Full activation of antigen-responding lymphocytes requires cooperating signals from the tumor necrosis factor (TNF)-related cytokines and their specific receptors. LIGHT, a lymphotoxin-beta (LTbeta)-related TNF family member, modulates T-cell activation through two receptors, the herpesvirus entry mediator (HVEM) and indirectly through the LT-beta receptor. An unexpected finding revealed a non-canonical binding site on HVEM for the immunoglobulin superfamily member, B and T lymphocyte attenuator (BTLA), and an inhibitory signaling protein suppressing T-cell activation. Thus, HVEM can act as a molecular switch between proinflammatory and inhibitory signaling. The non-canonical HVEM-BTLA pathway also acts to counter LTbetaR signaling that promotes the proliferation of antigen-presenting dendritic cells (DCs) within lymphoid tissue microenvironments. These results indicate LTbeta receptor and HVEM-BTLA pathways form an integrated signaling circuit. Targeting these cytokine pathways with specific antagonists (antibody or decoy receptor) can alter lymphocyte differentiation and activation. Alternately, agonists directed at their cell surface receptors can restore homeostasis and potentially reset immune and inflammatory processes, which may be useful in treating autoimmune and infectious diseases and cancer.
Collapse
Affiliation(s)
- Carl F Ware
- Division of Molecular Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.
| |
Collapse
|
42
|
Neo-lymphoid aggregates in the adult liver can initiate potent cell-mediated immunity. PLoS Biol 2009; 7:e1000109. [PMID: 19468301 PMCID: PMC2680335 DOI: 10.1371/journal.pbio.1000109] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Accepted: 03/27/2009] [Indexed: 12/18/2022] Open
Abstract
Are lymph nodes really essential for successful immunizations? We found that the liver can compensate for missing lymphoid structures in initiating cellular, but not antibody-mediated, immunity. Subcutaneous immunization delivers antigen (Ag) to local Ag-presenting cells that subsequently migrate into draining lymph nodes (LNs). There, they initiate the activation and expansion of lymphocytes specific for their cognate Ag. In mammals, the structural environment of secondary lymphoid tissues (SLTs) is considered essential for the initiation of adaptive immunity. Nevertheless, cold-blooded vertebrates can initiate potent systemic immune responses even though they lack conventional SLTs. The emergence of lymph nodes provided mammals with drastically improved affinity maturation of B cells. Here, we combine the use of different strains of alymphoplastic mice and T cell migration mutants with an experimental paradigm in which the site of Ag delivery is distant from the site of priming and inflammation. We demonstrate that in mammals, SLTs serve primarily B cell priming and affinity maturation, whereas the induction of T cell-driven immune responses can occur outside of SLTs. We found that mice lacking conventional SLTs generate productive systemic CD4- as well as CD8-mediated responses, even under conditions in which draining LNs are considered compulsory for the initiation of adaptive immunity. We describe an alternative pathway for the induction of cell-mediated immunity (CMI), in which Ag-presenting cells sample Ag and migrate into the liver where they induce neo-lymphoid aggregates. These structures are insufficient to support antibody affinity maturation and class switching, but provide a novel surrogate environment for the initiation of CMI. Lymph nodes (LNs) are believed to be the most important tissues initiating immune responses by facilitating the activation of T and B lymphocytes. Mice lacking such LNs (called alymphoplastic) are severely immune compromised and resistant to immunizations. We discovered that the immune-deficiency of such alymphoplastic mice is actually not caused by the loss of LNs, but rather by the underlying genetic lesion. Surprisingly, mice lacking all lymph nodes can still mount potent T cell-mediated immune responses. We also discovered that T and B cells have completely different structural requirements for their activation/maturation. Whereas B cells rely on LNs to become efficient antibody-producing cells, T cells can be activated successfully outside of such dedicated tissues. So—in the absence of LNs—antigens delivered by immunization are actively transported into the liver where cellular immunity is initiated. The mammalian fetal liver is responsible for the early formation of blood and immune cells, and we propose that the adult liver can still provide a niche for T cell–antigen encounters. During evolution, T and B cells emerged simultaneously, allowing cold-blooded vertebrates (which lack LNs) to launch adaptive immune responses. The development of LNs in mammals coincided with a drastic improvement in antibody affinity maturation, whereas T cells remain LN-independent to this day.
Collapse
|
43
|
Hagge DA, Saunders BM, Ebenezer GJ, Ray NA, Marks VT, Britton WJ, Krahenbuhl JL, Adams LB. Lymphotoxin-alpha and TNF have essential but independent roles in the evolution of the granulomatous response in experimental leprosy. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:1379-89. [PMID: 19246648 DOI: 10.2353/ajpath.2009.080550] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Recent studies identified an association between genetic variants in the lymphotoxin-alpha (LTalpha) gene and leprosy. To study the influence of LTalpha on the control of experimental leprosy, both low- and high-dose Mycobacterium leprae foot pad (FP) infections were evaluated in LTalpha-deficient chimeric (cLTalpha(-/-)) and control chimeric (cB6) mice. Cellular responses to low-dose infection in cLTalpha(-/-) mice were dramatically different, with reduced accumulation of CD4(+) and CD8(+) lymphocytes and macrophages and failure to form granulomas. Growth of M. leprae was contained for 6 months, but augmented late in infection. In contrast, tumor necrosis factor knockout and tumor necrosis factor receptor 1 knockout FPs exhibited extensive inflammatory infiltration with an increase in M. leprae growth throughout infection. Following high-dose infection, cB6 FP induration peaked at 4 weeks and was maintained for 12 weeks. Induration was not sustained in cLTalpha(-/-) FPs that contained few lymphocytes and no granulomas. There was a reduction in the expression levels of inflammatory cytokines, chemokines, and chemokine receptors, including nitric oxide synthase 2, vascular cell adhesion molecule, and intercellular cell adhesion molecule. Furthermore, cLTalpha(-/-) popliteal lymph nodes contained a higher proportion of naïve CD44(lo)CD62L(hi) T cells than cB6 mice, suggestive of reduced T cell activation. Therefore, both LTalpha and tumor necrosis factor are essential for the regulation of the granuloma, but they have distinctive roles in the recruitment of lymphocytes and maintenance of the granulomatous response during chronic M. leprae infection.
Collapse
Affiliation(s)
- Deanna A Hagge
- DHHS, HRSA, Bureau of Primary Health Care, National Hansen's Disease Programs Laboratory Research Branch at LSU-SVM, Skip Bertman Drive, Baton Rouge, LA 70803, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Heer AK, Harris NL, Kopf M, Marsland BJ. CD4+and CD8+T Cells Exhibit Differential Requirements for CCR7-Mediated Antigen Transport during Influenza Infection. THE JOURNAL OF IMMUNOLOGY 2008; 181:6984-94. [DOI: 10.4049/jimmunol.181.10.6984] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
45
|
Kursar M, Jänner N, Pfeffer K, Brinkmann V, Kaufmann SHE, Mittrücker HW. Requirement of secondary lymphoid tissues for the induction of primary and secondary T cell responses against Listeria monocytogenes. Eur J Immunol 2008; 38:127-38. [PMID: 18050270 DOI: 10.1002/eji.200737142] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Activation of naive T cells is tightly controlled and depends on cognate interactions with professional antigen-presenting cells. We analyzed dependency on secondary lymphoid tissues for the activation of naive and memory CD4(+) and CD8(+) T cells following primary and secondary Listeria monocytogenes infection, respectively. In splenectomized lymphotoxin-beta receptor-deficient mice, lacking all secondary lymphoid tissues, oral infection with L. monocytogenes failed to induce bacteria-specific CD4(+) and CD8(+) T cell responses. Treatment of splenectomized wild-type mice with FTY720, a drug that prevents egress of T cells from lymph nodes, also reduced T cell responses after oral L. monocytogenes infection and blocked T cell responses after intravenous infection. FTY720-treated wild-type and lymphotoxin-beta receptor-deficient mice show only slightly impaired recall responses. However, T cell responses were profoundly inhibited when mice were splenectomized subsequently to recovery from primary infection. T cell transfer experiments demonstrated that the impaired secondary T cell response was not simply due to removal of a large fraction of memory T cells by splenectomy. Overall, these results indicate that not only primary T cell responses, but also secondary T cell responses, highly depend on the lymphoid environment for effective activation.
Collapse
Affiliation(s)
- Mischo Kursar
- Max-Planck-Institute for Infection Biology, Department of Immunology, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
46
|
De Trez C, Schneider K, Potter K, Droin N, Fulton J, Norris PS, Ha SW, Fu YX, Murphy T, Murphy KM, Pfeffer K, Benedict CA, Ware CF. The inhibitory HVEM-BTLA pathway counter regulates lymphotoxin receptor signaling to achieve homeostasis of dendritic cells. THE JOURNAL OF IMMUNOLOGY 2008; 180:238-48. [PMID: 18097025 DOI: 10.4049/jimmunol.180.1.238] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Proliferation of dendritic cells (DC) in the spleen is regulated by positive growth signals through the lymphotoxin (LT)-beta receptor; however, the countering inhibitory signals that achieve homeostatic control are unresolved. Mice deficient in LTalpha, LTbeta, LTbetaR, and the NFkappaB inducing kinase show a specific loss of CD8- DC subsets. In contrast, the CD8alpha- DC subsets were overpopulated in mice deficient in the herpesvirus entry mediator (HVEM) or B and T lymphocyte attenuator (BTLA). HVEM- and BTLA-deficient DC subsets displayed a specific growth advantage in repopulating the spleen in competitive replacement bone marrow chimeric mice. Expression of HVEM and BTLA were required in DC and in the surrounding microenvironment, although DC expression of LTbetaR was necessary to maintain homeostasis. Moreover, enforced activation of the LTbetaR with an agonist Ab drove expansion of CD8alpha- DC subsets, overriding regulation by the HVEM-BTLA pathway. These results indicate the HVEM-BTLA pathway provides an inhibitory checkpoint for DC homeostasis in lymphoid tissue. Together, the LTbetaR and HVEM-BTLA pathways form an integrated signaling network regulating DC homeostasis.
Collapse
Affiliation(s)
- Carl De Trez
- Division of Molecular Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Carragher DM, Rangel-Moreno J, Randall TD. Ectopic lymphoid tissues and local immunity. Semin Immunol 2008; 20:26-42. [PMID: 18243731 DOI: 10.1016/j.smim.2007.12.004] [Citation(s) in RCA: 210] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2007] [Accepted: 12/06/2007] [Indexed: 01/28/2023]
Abstract
Ectopic or tertiary lymphoid tissues develop at sites of inflammation or infection in peripheral, non-lymphoid organs. These tissues are architecturally similar to conventional secondary lymphoid organs, with separated B and T cell areas, specialized populations of dendritic cells, well-differentiated stromal cells and high endothelial venules. Ectopic lymphoid tissues are often associated with the local pathology that results from chronic infection or chronic inflammation. However, there are also examples in which ectopic lymphoid tissues appear to contribute to local protective immune responses. Here we review how ectopic lymphoid structures develop and function in the context of local immunity and pathology.
Collapse
Affiliation(s)
- Damian M Carragher
- Trudeau Institute, 154 Algonquin Avenue, Saranac Lake, NY 12983, United States
| | | | | |
Collapse
|
48
|
Schrama D, Voigt H, Eggert AO, Xiang R, Zhou H, Schumacher TNM, Andersen MH, thor Straten P, Reisfeld RA, Becker JC. Immunological tumor destruction in a murine melanoma model by targeted LTalpha independent of secondary lymphoid tissue. Cancer Immunol Immunother 2008; 57:85-95. [PMID: 17605009 PMCID: PMC11030041 DOI: 10.1007/s00262-007-0352-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2006] [Accepted: 05/26/2007] [Indexed: 02/04/2023]
Abstract
BACKGROUND We previously demonstrated that targeting lymphotoxin alpha (LTalpha) to the tumor evokes its immunological destruction in a syngeneic B16 melanoma model. Since treatment was associated with the induction of peritumoral tertiary lymphoid tissue, we speculated that the induced immune response was initiated at the tumor site. METHODS AND RESULTS In order to directly test this notion, we analyzed the efficacy of tumor targeted LTalpha in LTalpha knock-out (LTalpha(-/-)) mice which lack peripheral lymph nodes. To this end, we demonstrate that tumor-targeted LTalpha mediates the induction of specific T-cell responses even in the absence of secondary lymphoid organs. In addition, this effect is accompanied by the initiation of tertiary lymphoid tissue at the tumor site in which B and T lymphocytes are compartmentalized in defined areas and which harbor expanded numbers of tumor specific T cells as demonstrated by in situ TRP-2/K(b) tetramer staining. Mechanistically, targeted LTalpha therapy seems to induce changes at the tumor site which allows a coordinated interaction of immune competent cells triggering the induction of tertiary lymphoid tissue. CONCLUSION Thus, our data demonstrate that targeted LTalpha promotes an accelerated immune response by enabling the priming of T cells at the tumor site.
Collapse
Affiliation(s)
- David Schrama
- Department of Dermatology, Julius-Maximilians-University, University of Würzburg, Josef-Schneider-Str. 2, Buildg. D8, 97080, Würzburg, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Moyron-Quiroz J, Rangel-Moreno J, Carragher DM, Randall TD. The function of local lymphoid tissues in pulmonary immune responses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 590:55-68. [PMID: 17191377 DOI: 10.1007/978-0-387-34814-8_4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
50
|
Oxelius VA. Personal glimpses of Robert A. Good. Immunol Res 2007. [DOI: 10.1007/s12026-007-0008-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|