1
|
Zhu Y, Wei L, Zwygart ACA, Gaínza P, Khac QO, Olgiati F, Kurum A, Tang L, Correia B, Tapparel C, Stellacci F. A Synthetic Multivalent Lipopeptide Derived from Pam3CSK4 with Irreversible Influenza Inhibition and Immuno-Stimulating Effects. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307709. [PMID: 38438885 DOI: 10.1002/smll.202307709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 02/23/2024] [Indexed: 03/06/2024]
Abstract
The activation of the host adaptive immune system is crucial for eliminating viruses. However, influenza infection often suppresses the innate immune response that precedes adaptive immunity, and the adaptive immune responses are typically delayed. Dendritic cells, serving as professional antigen-presenting cells, have a vital role in initiating the adaptive immune response. In this study, an immuno-stimulating antiviral system (ISAS) is introduced, which is composed of the immuno-stimulating adjuvant lipopeptide Pam3CSK4 that acts as a scaffold onto which it is covalently bound 3 to 4 influenza-inhibiting peptides. The multivalent display of peptides on the scaffold leads to a potent inhibition against H1N1 (EC50 = 20 nM). Importantly, the resulting lipopeptide, Pam3FDA, shows an irreversible inhibition mechanism. The chemical modification of peptides on the scaffold maintains Pam3CSK4's ability to stimulate dendritic cell maturation, thereby rendering Pam3FDA a unique antiviral. This is attributed to its immune activation capability, which also acts in synergy to expedite viral elimination.
Collapse
Affiliation(s)
- Yong Zhu
- Institute of Materials, École Polytechnique Fédérale de Lausanne Station 12, Lausanne, CH-1015, Switzerland
| | - Lixia Wei
- Institute of Materials, École Polytechnique Fédérale de Lausanne Station 12, Lausanne, CH-1015, Switzerland
| | - Arnaud Charles-Antoine Zwygart
- Department of Microbiology and Molecular Medicine, University of Geneva, CMU Rue Michel-Servet 1, Geneva 4, CH-1211, Switzerland
| | - Pablo Gaínza
- Interschool Institute of Bioengineering, École Polytechnique Fédérale de Lausanne Station 12, Lausanne, CH-1015, Switzerland
| | - Quy Ong Khac
- Institute of Materials, École Polytechnique Fédérale de Lausanne Station 12, Lausanne, CH-1015, Switzerland
| | - Francesca Olgiati
- Institute of Materials, École Polytechnique Fédérale de Lausanne Station 12, Lausanne, CH-1015, Switzerland
| | - Armand Kurum
- Interschool Institute of Bioengineering, École Polytechnique Fédérale de Lausanne Station 12, Lausanne, CH-1015, Switzerland
| | - Li Tang
- Interschool Institute of Bioengineering, École Polytechnique Fédérale de Lausanne Station 12, Lausanne, CH-1015, Switzerland
| | - Bruno Correia
- Interschool Institute of Bioengineering, École Polytechnique Fédérale de Lausanne Station 12, Lausanne, CH-1015, Switzerland
| | - Caroline Tapparel
- Department of Microbiology and Molecular Medicine, University of Geneva, CMU Rue Michel-Servet 1, Geneva 4, CH-1211, Switzerland
| | - Francesco Stellacci
- Institute of Materials, École Polytechnique Fédérale de Lausanne Station 12, Lausanne, CH-1015, Switzerland
| |
Collapse
|
2
|
Kobayashi T, Iijima K, Matsumoto K, Lama JK, Kita H. Lung-resident CD69 +ST2 + T H2 cells mediate long-term type 2 memory to inhaled antigen in mice. J Allergy Clin Immunol 2023; 152:167-181.e6. [PMID: 36720287 PMCID: PMC10330297 DOI: 10.1016/j.jaci.2023.01.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/30/2023]
Abstract
BACKGROUND Chronic airway diseases such as asthma are characterized by persistent type 2 immunity in the airways. We know little about the mechanisms that explain why type 2 inflammation continues in these diseases. OBJECTIVE We used mouse models to investigate the mechanisms involved in long-lasting immune memory. METHODS Naive mice were exposed intranasally to ovalbumin (OVA) antigen with Alternaria extract as an adjuvant. Type 2 memory was analyzed by parabiosis model, flow cytometry with in vivo antibody labeling, and intranasal OVA recall challenge. Gene-deficient mice were used to analyze the mechanisms. RESULTS In the parabiosis model, mice previously exposed intranasally to OVA with Alternaria showed more robust antigen-specific immune responses and airway inflammation than mice with circulating OVA-specific T cells. After a single airway exposure to OVA with Alternaria, CD69+ST2+ TH2-type T cells, which highly express type 2 cytokine messenger RNA and lack CD62L expression, appeared in lung tissue within 5 days and persisted for at least 84 days. When exposed again to OVA in vivo, these cells produced type 2 cytokines quickly without involving circulating T cells. Development of tissue-resident CD69+ST2+ TH2 cells and long-term memory to an inhaled antigen were abrogated in mice deficient in ST2 or IL-33, but not TSLP receptor. CONCLUSION CD69+ST2+ TH2 memory cells develop quickly in lung tissue after initial allergen exposure and persist for a prolonged period. The ST2/IL-33 pathway may play a role in the development of immune memory in lung to certain allergens.
Collapse
Affiliation(s)
- Takao Kobayashi
- Division of Allergic Diseases, Asthma and Clinical Immunology, and Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Koji Iijima
- Division of Allergic Diseases, Asthma and Clinical Immunology, and Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Koji Matsumoto
- Division of Allergic Diseases, Asthma and Clinical Immunology, and Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Jyoti K Lama
- Immunology Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester and Scottsdale, Rochester, Minn
| | - Hirohito Kita
- Division of Allergic Diseases, Asthma and Clinical Immunology, and Department of Medicine, Mayo Clinic, Scottsdale, Ariz; Department of Immunology, Mayo Clinic, Rochester, and Mayo Clinic, Scottsdale, Ariz.
| |
Collapse
|
3
|
How dendritic cells sense and respond to viral infections. Clin Sci (Lond) 2021; 135:2217-2242. [PMID: 34623425 DOI: 10.1042/cs20210577] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/15/2021] [Accepted: 09/23/2021] [Indexed: 12/26/2022]
Abstract
The ability of dendritic cells (DCs) to sense viral pathogens and orchestrate a proper immune response makes them one of the key players in antiviral immunity. Different DC subsets have complementing functions during viral infections, some specialize in antigen presentation and cross-presentation and others in the production of cytokines with antiviral activity, such as type I interferons. In this review, we summarize the latest updates concerning the role of DCs in viral infections, with particular focus on the complex interplay between DC subsets and severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Despite being initiated by a vast array of immune receptors, DC-mediated antiviral responses often converge towards the same endpoint, that is the production of proinflammatory cytokines and the activation of an adaptive immune response. Nonetheless, the inherent migratory properties of DCs make them a double-edged sword and often viral recognition by DCs results in further viral dissemination. Here we illustrate these various aspects of the antiviral functions of DCs and also provide a brief overview of novel antiviral vaccination strategies based on DCs targeting.
Collapse
|
4
|
Blockade of CD40L inhibits immunogenic maturation of lung dendritic cells: Implications for the role of lung iNKT cells in mouse models of asthma. Mol Immunol 2020; 121:167-185. [PMID: 32229377 DOI: 10.1016/j.molimm.2020.03.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 03/15/2020] [Accepted: 03/15/2020] [Indexed: 12/15/2022]
Abstract
Some studies have shown that maturation of dendritic cells (DCs) is modulated directly by pathogen components via pattern recognition receptors such as Toll-like receptors, but also by signal like CD40 ligand (CD40 L or CD154) mediated by activated T cells. Several reports indicate that invariant natural killer T (iNKT) cells up-regulate CD40 L upon stimulation and thereby induce activation and maturation of DCs through crosslink with CD40. Our previous findings indicated that iNKT cells promote Th2 cell responses through the induction of immunogenic maturation of lung DCs (LDCs) in the asthmatic murine, but its mechanism remains unclear. Therefore, we investigated the immunomodulatory effects of blockade of CD40 L using anti-CD40 L treatment on Th2 cell responses and immunogenic maturation of LDCs, and further analyzed whether these influences of blockade of CD40 L were related to lung iNKT cells using iNKT cell-deficient mice and the combination treatment of specific iNKT cell activation with anti-CD40 L treatment in murine models of asthma. Our findings showed that blockade of CD40 L using anti-CD40 L treatment attenuated Th2 cell responses in wild-type (WT) mice, but not in CD1d-deficient mice sensitized and challenged with ovalbumin (OVA) or house dust mite (HDM). Meanwhile, blockade of CD40 L down-regulated immunogenic maturation of LDCs in WT mice, but not in CD1d-deficient mice sensitized and challenged with OVA. Additionally, agonistic anti-CD40 treatment reversed the inhibitory effects of anti-CD40 L treatment on Th2 cell responses and LDC activation in an OVA-induced mouse model of asthma. Furthermore, LDCs from asthmatic mice treated with anti-CD40 L could significantly reduce the influence on Th2 cell responses in vivo and in vitro. Finally, α-Galactosylceramide plus anti-CD40 L treatment stimulated lung iNKT cells, but suppressed Th2 cell responses in the asthmatic mice. Taken together, our data raise an evidence that blockade of CD40 L attenuates Th2 cell responses through the inhibition of immunogenic maturation of LDCs, which may be at least partially related to lung iNKT cells in murine models of asthma.
Collapse
|
5
|
Palomino-Segura M, Perez L, Farsakoglu Y, Virgilio T, Latino I, D'Antuono R, Chatziandreou N, Pizzagalli DU, Wang G, García-Sastre A, Sallusto F, Carroll MC, Neyrolles O, Gonzalez SF. Protection against influenza infection requires early recognition by inflammatory dendritic cells through C-type lectin receptor SIGN-R1. Nat Microbiol 2019; 4:1930-1940. [PMID: 31358982 PMCID: PMC6817362 DOI: 10.1038/s41564-019-0506-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 06/06/2019] [Indexed: 12/23/2022]
Abstract
The early phase of influenza infection occurs in the upper respiratory tract and the trachea, but little is known about the initial events of virus recognition and control of viral dissemination by the immune system. Here, we report that inflammatory dendritic cells (IDCs) are recruited to the trachea shortly after influenza infection through type I interferon-mediated production of the chemokine CCL2. We further show that recruited IDCs express the C-type lectin receptor SIGN-R1, which mediates direct recognition of the virus by interacting with N-linked glycans present in glycoproteins of the virion envelope. Activation of IDCs via SIGN-R1 triggers the production of the chemokines CCL5, CXCL9 and CXCL10, which initiate the recruitment of protective natural killer (NK) cells in the infected trachea. In the absence of SIGN-R1, the recruitment and activation of NK cells is impaired, leading to uncontrolled viral proliferation. In sum, our results provide insight into the orchestration of the early cellular and molecular events involved in immune protection against influenza.
Collapse
Affiliation(s)
- Miguel Palomino-Segura
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Graduate School of Cellular and Molecular Sciences, Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Laurent Perez
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Yagmur Farsakoglu
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Graduate School of Cellular and Molecular Sciences, Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Tommaso Virgilio
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Graduate School of Cellular and Molecular Sciences, Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Irene Latino
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Rocco D'Antuono
- Light Microscopy STP, The Francis Crick Institute, London, UK
| | - Nikolaos Chatziandreou
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Diego U Pizzagalli
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Computational Science, Università della Svizzera italiana, Lugano, Switzerland
| | - Guojun Wang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogen Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogen Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute for Microbiology, ETH Zurich, Zurich, Switzerland
| | - Michael C Carroll
- Program in Cellular and Molecular Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse CNRS, UPS, Toulouse, France
| | - Santiago F Gonzalez
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland.
| |
Collapse
|
6
|
Loghmani A, Raoofi R, Ownagh A, Delirezh N. Alternaria alternata acts on human Monocyte-derived Dendritic cells to mediate Th2/Th17 polarisation. Allergol Immunopathol (Madr) 2017; 45:175-182. [PMID: 27823917 DOI: 10.1016/j.aller.2016.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/05/2016] [Accepted: 07/08/2016] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Although the mechanism of asthma is not precisely understood in humans, clinical and epidemiological studies have offered a potential relationship between exposure to environmental fungi, such as Alternaria alternata (A. alternata) and the development and exacerbation of asthma. The aim of this project is to investigate the mechanisms of Th2 responses by A. alternata as a clinically relevant model for the environmental exposure. MATERIALS AND METHODS Plastic adherent monocytes were cultured with granulocyte macrophage colony stimulating factor (GM-CSF) and interleukin-4 (IL-4) to convert these cells into Monocyte-derived Dendritic cells (MoDc) and then matured in the presence of Monocyte-Conditioned Medium (MCM) as the control group and MCM+ A. alternata extract as the inductive groups. RESULTS The results indicated that the expression of CD14 decreased and CD83 and anti-human leukocyte antigen-DR (HLA-DR) increased in the inductive groups in comparison with the control group. More importantly, A. alternata inhibited IL-12 production by activated dendritic cells (DCs), and the DCs exposed to A. alternata enhanced the Th2 polarisation of CD4+ T cells. The production amount of IL-10 overcame IL-12 as well as Il-23 increased significantly, and hand in T cells the production of cytokines Interferon-γ (IFN-γ) decreased. However, both IL-17 and IL-4 increased (p<0.05). Phagocytic activity in the inductive groups decreased significantly compared with the control group. CONCLUSION The asthma-related environmental fungus A. alternata, with an effect on dendritic cells profile mediates TH2/TH17. Such immunodysregulation properties of causative environmental fungi may explain their strong relationship with human asthma and allergic diseases.
Collapse
Affiliation(s)
- A Loghmani
- Department of Microbiology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran.
| | - R Raoofi
- Department of Infectious Diseases, University of Jahrom Medical Science, Jahrom, Iran
| | - A Ownagh
- Department of Infectious Diseases, University of Jahrom Medical Science, Jahrom, Iran
| | - N Delirezh
- Department of Infectious Diseases, University of Jahrom Medical Science, Jahrom, Iran
| |
Collapse
|
7
|
Daniels NJ, Hyde E, Ghosh S, Seo K, Price KM, Hoshino K, Kaisho T, Okada T, Ronchese F. Antigen-specific cytotoxic T lymphocytes target airway CD103+ and CD11b+ dendritic cells to suppress allergic inflammation. Mucosal Immunol 2016; 9:229-39. [PMID: 26104914 DOI: 10.1038/mi.2015.55] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/19/2015] [Indexed: 02/04/2023]
Abstract
Allergic airway inflammation is driven by the recognition of inhaled allergen by T helper type 2 (Th2) cells in the airway and lung. Allergen-specific cytotoxic T lymphocytes (CTLs) can strongly reduce airway inflammation, however, the mechanism of their inhibitory activity is not fully defined. We used mouse models to show that allergen-specific CTLs reduced early cytokine production by Th2 cells in lung, and their subsequent accumulation and production of interleukin (IL)-4 and IL-13. In addition, treatment with specific CTLs also increased the proportion of caspase(+) dendritic cells (DCs) in mediastinal lymph node (MLN), and decreased the numbers of CD103(+) and CD11b(+) DCs in the lung. This decrease required expression of the cytotoxic mediator perforin in CTLs and of the appropriate MHC-antigen ligand on DCs, suggesting that direct CTL-DC contact was necessary. Lastly, lung imaging experiments revealed that in airway-challenged mice XCR1-GFP(+) DCs, corresponding to the CD103(+) DC subset, and XCR1-GFP(-) CD11c(+) cells, which include CD11b(+) DCs and alveolar macrophages, both clustered in the areas surrounding the small airways and were closely associated with allergen-specific CTLs. Thus, allergen-specific CTLs reduce allergic airway inflammation by depleting CD103(+) and CD11b(+) DC populations in the lung, and may constitute a mechanism through which allergic immune responses are regulated.
Collapse
Affiliation(s)
- N J Daniels
- Malaghan Institute of Medical Research, Wellington, New Zealand.,University of Otago, Wellington, New Zealand
| | - E Hyde
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - S Ghosh
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - K Seo
- Lab for Tissue Dynamics, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), Yokohama, Japan
| | - K M Price
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - K Hoshino
- Laboratory for Inflammatory Regulation, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), Yokohama, Japan.,Department of Immunology, Faculty of Medicine, Kagawa University, Kita-gun, Japan
| | - T Kaisho
- Laboratory for Inflammatory Regulation, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), Yokohama, Japan.,Laboratory for Immune Regulation, World Premier International Immunology Frontier Research Center, Osaka University Suita, Osaka, Japan.,Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - T Okada
- Lab for Tissue Dynamics, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), Yokohama, Japan.,PRESTO, Japan Science and Technology Agency, Saitama, Japan.,Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - F Ronchese
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
8
|
Chicken bone marrow-derived dendritic cells maturation in response to infectious bursal disease virus. Vet Immunol Immunopathol 2015; 164:51-5. [PMID: 25613777 DOI: 10.1016/j.vetimm.2014.12.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Revised: 12/11/2014] [Accepted: 12/31/2014] [Indexed: 01/06/2023]
Abstract
Infectious bursal disease virus (IBDV) is highly contagious disease which easily lead to immunosuppression and a decreased response to vaccinations in young chicken. Since dendritic cells (DCs) are crucial to induce immunity and their maturation and functions are influenced by microbial and environmental stimuli, we investigated the effects of inactivated IBDV and IBDV on chicken DC activation and maturation. Chicken bone marrow-derived dendritic cells (chBM-DCs) cultured in complete medium (including recombinant chicken: granulocyte-macrophage colony-stimulating factor and interleukin 4) expressed high levels of MHC-II and the putative CD11c. After LPS or virus stimulation, chBM-DCs displayed the typical morphology of DCs. In addition, stimulation by LPS or viruses significantly elevated chBM-DCs surface expression levels of CD40 and CD86 molecules, as well as the ability to induce T-cell proliferative response, compared to the non-stimulated chBM-DCs. Interestingly, inactive IBDV showed stronger ability to up-regulate expression levels of CD40 and CD86 molecules and stimulate naive T cells proliferation than live IBDV. These results revealed that live viruses infection impaired DC maturation and functions, probably explaining why chickens infected with IBDV fails to trigger an effective specific immune response or develop immune memory.
Collapse
|
9
|
Altenburg AF, Rimmelzwaan GF, de Vries RD. Virus-specific T cells as correlate of (cross-)protective immunity against influenza. Vaccine 2015; 33:500-6. [DOI: 10.1016/j.vaccine.2014.11.054] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 10/30/2014] [Accepted: 11/28/2014] [Indexed: 12/12/2022]
|
10
|
Persistent and compartmentalised disruption of dendritic cell subpopulations in the lung following influenza A virus infection. PLoS One 2014; 9:e111520. [PMID: 25398128 PMCID: PMC4232261 DOI: 10.1371/journal.pone.0111520] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 09/25/2014] [Indexed: 11/19/2022] Open
Abstract
Immunological homeostasis in the respiratory tract is thought to require balanced interactions between networks of dendritic cell (DC) subsets in lung microenvironments in order to regulate tolerance or immunity to inhaled antigens and pathogens. Influenza A virus (IAV) poses a serious threat of long-term disruption to this balance through its potent pro-inflammatory activities. In this study, we have used a BALB/c mouse model of A/PR8/34 H1N1 Influenza Type A Virus infection to examine the effects of IAV on respiratory tissue DC subsets during the recovery phase following clearance of the virus. In adult mice, we found differences in the kinetics and activation states of DC residing in the airway mucosa (AMDC) compared to those in the parenchymal lung (PLDC) compartments. A significant depletion in the percentage of AMDC was observed at day 4 post-infection that was associated with a change in steady-state CD11b+ and CD11b- AMDC subset frequencies and significantly elevated CD40 and CD80 expression and that returned to baseline by day 14 post-infection. In contrast, percentages and total numbers of PLDC were significantly elevated at day 14 and remained so until day 21 post-infection. Accompanying this was a change in CD11b+and CD11b- PLDC subset frequencies and significant increase in CD40 and CD80 expression at these time points. Furthermore, mice infected with IAV at 4 weeks of age showed a significant increase in total numbers of PLDC, and increased CD40 expression on both AMDC and PLDC, when analysed as adults 35 days later. These data suggest that the rate of recovery of DC populations following IAV infection differs in the mucosal and parenchymal compartments of the lung and that DC populations can remain disrupted and activated for a prolonged period following viral clearance, into adulthood if infection occurred early in life.
Collapse
|
11
|
Vanders RL, Murphy VE, Gibson PG, Hansbro PM, Wark PAB. CD8 T cells and dendritic cells: key players in the attenuated maternal immune response to influenza infection. J Reprod Immunol 2014; 107:1-9. [PMID: 25453203 DOI: 10.1016/j.jri.2014.09.051] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 08/01/2014] [Accepted: 09/09/2014] [Indexed: 12/16/2022]
Abstract
Pregnancy provides a unique challenge for maternal immunity, requiring the ability to tolerate the presence of a semi-allogeneic foetus, and yet still being capable of inducing an immune response against invading pathogens. To achieve this, numerous changes must occur in the activity and function of maternal immune cells throughout the course of pregnancy. Respiratory viruses take advantage of these changes, altering the sensitive balance of maternal immunity, leaving the mother with increased susceptibility to viral infections and increased disease severity. Influenza virus is one of the most common respiratory virus infections during pregnancy, leading to an increased risk of ICU hospitalisations, pneumonia, acute respiratory distress syndrome and even death. Whilst much research has been performed to understand the changes that must take place in maternal immunity during pregnancy, considerable work is still needed to fully comprehend this tremendous feat. To date, few studies have focused on the alterations that occur in maternal immunity during respiratory virus infections. This review highlights the role of dendritic cells (DCs) and CD8 T cells during pregnancy, and the changes that occur in these antiviral cells following influenza virus infections.
Collapse
Affiliation(s)
- Rebecca L Vanders
- Centre for Asthma and Respiratory Diseases, The University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia.
| | - Vanessa E Murphy
- Centre for Asthma and Respiratory Diseases, The University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia.
| | - Peter G Gibson
- Centre for Asthma and Respiratory Diseases, The University of Newcastle, Newcastle, NSW, Australia; The Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia.
| | - Philip M Hansbro
- Centre for Asthma and Respiratory Diseases, The University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia.
| | - Peter A B Wark
- Centre for Asthma and Respiratory Diseases, The University of Newcastle, Newcastle, NSW, Australia; The Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia.
| |
Collapse
|
12
|
Yu CI, Becker C, Metang P, Marches F, Wang Y, Toshiyuki H, Banchereau J, Merad M, Palucka AK. Human CD141+ dendritic cells induce CD4+ T cells to produce type 2 cytokines. THE JOURNAL OF IMMUNOLOGY 2014; 193:4335-43. [PMID: 25246496 DOI: 10.4049/jimmunol.1401159] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Dendritic cells (DCs) play the central role in the priming of naive T cells and the differentiation of unique effector T cells. In this study, using lung tissues and blood from both humans and humanized mice, we analyzed the response of human CD1c(+) and CD141(+) DC subsets to live-attenuated influenza virus. Specifically, we analyzed the type of CD4(+) T cell immunity elicited by live-attenuated influenza virus-exposed DCs. Both DC subsets induce proliferation of allogeneic naive CD4(+) T cells with the capacity to secrete IFN-γ. However, CD141(+) DCs are uniquely able to induce the differentiation of IL-4- and IL-13-producing CD4(+) T cells. CD141(+) DCs induce IL-4- and IL-13-secreting CD4(+) T cells through OX40 ligand. Thus, CD141(+) DCs demonstrate remarkable plasticity in guiding adaptive immune responses.
Collapse
Affiliation(s)
- Chun I Yu
- Baylor Institute for Immunology Research, Dallas, TX 75204
| | - Christian Becker
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029; Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | - Patrick Metang
- Baylor Institute for Immunology Research, Dallas, TX 75204
| | | | - Yuanyuan Wang
- Baylor Institute for Immunology Research, Dallas, TX 75204; Institute of Biomedical Studies, Baylor University, Waco, TX 76798
| | - Hori Toshiyuki
- College of Life Sciences, Ritsumeikan University, Shiga 525-8577, Japan
| | | | - Miriam Merad
- Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029; Department of Oncological Sciences, Mount Sinai School of Medicine, New York, NY 10029
| | | |
Collapse
|
13
|
Hara K, Iijima K, Elias MK, Seno S, Tojima I, Kobayashi T, Kephart GM, Kurabayashi M, Kita H. Airway uric acid is a sensor of inhaled protease allergens and initiates type 2 immune responses in respiratory mucosa. THE JOURNAL OF IMMUNOLOGY 2014; 192:4032-42. [PMID: 24663677 DOI: 10.4049/jimmunol.1400110] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Although type 2 immune responses to environmental Ags are thought to play pivotal roles in asthma and allergic airway diseases, the immunological mechanisms that initiate the responses are largely unknown. Many allergens have biologic activities, including enzymatic activities and abilities to engage innate pattern-recognition receptors such as TLR4. In this article, we report that IL-33 and thymic stromal lymphopoietin were produced quickly in the lungs of naive mice exposed to cysteine proteases, such as bromelain and papain, as a model for allergens. IL-33 and thymic stromal lymphopoietin sensitized naive animals to an innocuous airway Ag OVA, which resulted in production of type 2 cytokines and IgE Ab, and eosinophilic airway inflammation when mice were challenged with the same Ag. Importantly, upon exposure to proteases, uric acid (UA) was rapidly released into the airway lumen, and removal of this endogenous UA by uricase prevented type 2 immune responses. UA promoted secretion of IL-33 by airway epithelial cells in vitro, and administration of UA into the airways of naive animals induced extracellular release of IL-33, followed by both innate and adaptive type 2 immune responses in vivo. Finally, a potent UA synthesis inhibitor, febuxostat, mitigated asthma phenotypes that were caused by repeated exposure to natural airborne allergens. These findings provide mechanistic insights into the development of type 2 immunity to airborne allergens and recognize airway UA as a key player that regulates the process in respiratory mucosa.
Collapse
Affiliation(s)
- Kenichiro Hara
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Sant AJ, Chaves FA, Krafcik FR, Lazarski CA, Menges P, Richards K, Weaver JM. Immunodominance in CD4 T-cell responses: implications for immune responses to influenza virus and for vaccine design. Expert Rev Vaccines 2014; 6:357-68. [PMID: 17542751 DOI: 10.1586/14760584.6.3.357] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
CD4 T cells play a primary role in regulating immune responses to pathogenic organisms and to vaccines. Antigen-specific CD4 T cells provide cognate help to B cells, a requisite event for immunoglobulin switch and affinity maturation of B cells that produce neutralizing antibodies and also provide help to cytotoxic CD8 T cells, critical for their expansion and persistence as memory cells. Finally, CD4 T cells may participate directly in pathogen clearance via cell-mediated cytotoxicity or through production of cytokines. Understanding the role of CD4 T-cell immunity to viruses and other pathogens, as well as evaluation of the efficacy of vaccines, requires insight into the specificity of CD4 T cells. This review focuses on the events within antigen-presenting cells that focus CD4 T cells toward a limited number of peptide antigens within the pathogen or vaccine. The molecular events are discussed in light of the special challenges that the influenza virus poses, owing to the high degree of genetic variability, unpredictable pathogenicity and the repeated encounters that human populations face with this highly infectious pathogenic organism.
Collapse
Affiliation(s)
- Andrea J Sant
- David H Smith Center for Vaccine Biology and Immunology, Aab Institute and Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA.
| | | | | | | | | | | | | |
Collapse
|
15
|
CD4 T cell help is limiting and selective during the primary B cell response to influenza virus infection. J Virol 2013; 88:314-24. [PMID: 24155379 DOI: 10.1128/jvi.02077-13] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Influenza virus vaccination strategies are focused upon the elicitation of protective antibody responses through administration of viral protein through either inactivated virions or live attenuated virus. Often overlooked in this strategy is the CD4 T cell response: how it develops into memory, and how it may support future primary B cell responses to heterologous infection. Through the utilization of a peptide-priming regimen, this study describes a strategy for developing CD4 T cell memory with the capacity to robustly expand in the lung-draining lymph node after live influenza virus infection. Not only were frequencies of antigen-specific CD4 T cells enhanced, but these cells also supported an accelerated primary B cell response to influenza virus-derived protein, evidenced by high anti-nucleoprotein (NP) serum antibody titers early, while there is still active viral replication ongoing in the lung. NP-specific antibody-secreting cells and heightened frequencies of germinal center B cells and follicular T helper cells were also readily detectable in the draining lymph node. Surprisingly, a boosted memory CD4 T cell response was not sufficient to provide intermolecular help for antibody responses. Our study demonstrates that CD4 T cell help is selective and limiting to the primary antibody response to influenza virus infection and that preemptive priming of CD4 T cell help can promote effective and rapid conversion of naive B cells to mature antibody-secreting cells.
Collapse
|
16
|
PC61 (anti-CD25) treatment inhibits influenza A virus-expanded regulatory T cells and severe lung pathology during a subsequent heterologous lymphocytic choriomeningitis virus infection. J Virol 2013; 87:12636-47. [PMID: 24049180 DOI: 10.1128/jvi.00936-13] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Prior immunity to influenza A virus (IAV) in mice changes the outcome to a subsequent lymphocytic choriomeningitis virus (LCMV) infection and can result in severe lung pathology, similar to that observed in patients that died of the 1918 H1N1 pandemic. This pathology is induced by IAV-specific memory CD8(+) T cells cross-reactive with LCMV. Here, we discovered that IAV-immune mice have enhanced CD4(+) Foxp3(+) T-regulatory (Treg) cells in their lungs, leading us to question whether a modulation in the normal balance of Treg and effector T-cell responses also contributes to enhancing lung pathology upon LCMV infection of IAV-immune mice. Treg cell and interleukin-10 (IL-10) levels remained elevated in the lungs and mediastinal lymph nodes (mLNs) throughout the acute LCMV response of IAV-immune mice. PC61 treatment, used to decrease Treg cell levels, did not change LCMV titers but resulted in a surprising decrease in lung pathology upon LCMV infection in IAV-immune but not in naive mice. Associated with this decrease in pathology was a retention of Treg in the mLN and an unexpected partial clonal exhaustion of LCMV-specific CD8(+) T-cell responses only in IAV-immune mice. PC61 treatment did not affect cross-reactive memory CD8(+) T-cell proliferation. These results suggest that in the absence of IAV-expanded Treg cells and in the presence of cross-reactive memory, the LCMV-specific response was overstimulated and became partially exhausted, resulting in a decreased effector response. These studies suggest that Treg cells generated during past infections can influence the characteristics of effector T-cell responses and immunopathology during subsequent heterologous infections. Thus, in humans with complex infection histories, PC61 treatment may lead to unexpected results.
Collapse
|
17
|
Guibas GV, Makris M, Papadopoulos NG. Key Regulators of Sensitization and Tolerance: GM-CSF, IL-10, TGF-β and the Notch Signaling Pathway in Adjuvant-Free Experimental Models of Respiratory Allergy. Int Rev Immunol 2013; 32:307-23. [DOI: 10.3109/08830185.2013.794457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
18
|
Flórido M, Grima MA, Gillis CM, Xia Y, Turner SJ, Triccas JA, Stambas J, Britton WJ. Influenza A Virus Infection Impairs Mycobacteria-Specific T Cell Responses and Mycobacterial Clearance in the Lung during Pulmonary Coinfection. THE JOURNAL OF IMMUNOLOGY 2013; 191:302-11. [DOI: 10.4049/jimmunol.1202824] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
19
|
Tanyi JL, Chu CS. Dendritic cell-based tumor vaccinations in epithelial ovarian cancer: a systematic review. Immunotherapy 2013; 4:995-1009. [PMID: 23148752 DOI: 10.2217/imt.12.100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
After decades of extensive research, epithelial ovarian cancer still remains a lethal disease. Multiple new studies have reported that the immune system plays a critical role in the growth and spread of ovarian carcinoma. This review summarizes the development of dendritic cell (DC) vaccinations specific for ovarian cancer. So far, DC-based vaccines have induced effective antitumor responses in animal models, but only limited results from human clinical trials are available. Although DC-based immunotherapy has proven to be clinically safe and efficient at inducing tumor-specific immune responses, its clear role in the therapy of ovarian cancer still needs to be clarified. The relatively disappointing low-response rates in early clinical trials point to the need for the development of more effective and personalized DC-based anticancer vaccines. This article reviews the basic mechanisms, limitations and future directions of DC-based anti-ovarian cancer vaccine development.
Collapse
Affiliation(s)
- Janos L Tanyi
- Division of Gynecologic Oncology, Department of Obstetrics & Gynecology, University of Pennsylvania Health System, Philadelphia, PA, USA
| | | |
Collapse
|
20
|
Abstract
Pulmonary dendritic cells (DCs) constantly sample the tissue and traffic inhaled antigens to the lung-draining lymph node where they normally orchestrate an appropriate immune response. The dynamic ability of these professional antigen-presenting cells to promote tolerance or immunity has been intensively studied by several groups, including ours. Distinct DC subsets in both lymphoid and non-lymphoid tissues have been described based on their surface molecule expression and location. Current efforts to unravel DC development and function are providing insight into the various roles each subset offers the immune system. Elucidating DC functions, particularly in the lung, may then allow use of the inherent ability of these cells for enhanced vaccine strategies and therapeutics for pulmonary infections and diseases.
Collapse
Affiliation(s)
- A. Nicole Desch
- Integrated Department of Immunology, University of Colorado School of Medicine, Denver, CO, USA
| | - Peter M. Henson
- Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
- Integrated Department of Immunology, University of Colorado School of Medicine, Denver, CO, USA
| | - Claudia V. Jakubzick
- Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA,
- Integrated Department of Immunology, University of Colorado School of Medicine, Denver, CO, USA
| |
Collapse
|
21
|
Guibas GV, Makris M, Spandou E, Priftis KN. Exposure of immunologically naive laboratory rodents to antigen via the airways. Where does tolerance stop and sensitization begin? Clin Exp Allergy 2012; 42:1552-65. [DOI: 10.1111/j.1365-2222.2012.03974.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
| | - M. Makris
- Allergy Unit; 2nd Department of dermatology and Venereology; Medical School; “Attikon” General University Hospital; Athens; Greece
| | - E. Spandou
- Laboratory of Experimental Physiology; Medical School; Aristotle University of Thessaloniki; Thessaloniki; Greece
| | - K. N. Priftis
- 3rd Department of Pediatrics; Medical School; “Attikon” General University Hospital; Athens; Greece
| |
Collapse
|
22
|
Tel J, van der Leun AM, Figdor CG, Torensma R, de Vries IJM. Harnessing human plasmacytoid dendritic cells as professional APCs. Cancer Immunol Immunother 2012; 61:1279-88. [PMID: 22294456 PMCID: PMC3401502 DOI: 10.1007/s00262-012-1210-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 01/19/2012] [Indexed: 12/23/2022]
Abstract
The plasmacytoid dendritic cell (pDC) constitutes a unique DC subset that links the innate and adaptive arm of the immune system. Whereas the unique capability of pDCs to produce large amounts of type I IFNs in response to pathogen recognition is generally accepted, their antigen-presenting function is often neglected since most studies on antigen presentation are aimed at other DC subsets. Recently, pDCs were demonstrated capable to present antigen leading to protective tumor immunity. In this review, we discuss how pDCs could be exploited in the fight against cancer by analyzing their capacity to capture, process and (cross-) present antigen.
Collapse
Affiliation(s)
- Jurjen Tel
- Department of Tumor Immunology, Radboud University Nijmegen Medical Centre and Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | | | | | | | | |
Collapse
|
23
|
Type I interferon induction during influenza virus infection increases susceptibility to secondary Streptococcus pneumoniae infection by negative regulation of γδ T cells. J Virol 2012; 86:12304-12. [PMID: 22951826 DOI: 10.1128/jvi.01269-12] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The majority of deaths following influenza virus infection result from secondary bacterial superinfection, most commonly caused by Streptococcus pneumoniae. Several models have been proposed to explain how primary respiratory viral infections exacerbate secondary bacterial disease, but the mechanistic explanations have been contradictory. In this study, mice were infected with S. pneumoniae at different days after primary influenza A (X31) virus infection. Our findings show that the induction of type I interferons (IFNs) during a primary nonlethal influenza virus infection is sufficient to promote a deadly S. pneumoniae secondary infection. Moreover, mice deficient in type I interferon receptor (IFNAR knockout [KO] mice) effectively cleared the secondary bacterial infection from their lungs, increased the recruitment of neutrophils, and demonstrated an enhanced innate expression of interleukin-17 (IL-17) relative to wild-type (WT) mice. Lung γδ T cells were responsible for almost all IL-17 production, and their function is compromised during secondary S. pneumoniae infection of WT but not IFNAR KO mice. Adoptive transfer of γδ T cells from IFNAR KO mice reduced the susceptibility to secondary S. pneumoniae infection in the lung of WT mice. Altogether, our study highlights the importance of type I interferon as a key master regulator that is exploited by opportunistic pathogens such as S. pneumoniae. Our findings may be utilized to design effective preventive and therapeutic strategies that may be beneficial for coinfected patients during influenza epidemics.
Collapse
|
24
|
Müller L, Jaspers I. Epithelial cells, the "switchboard" of respiratory immune defense responses: effects of air pollutants. Swiss Med Wkly 2012; 142:w13653. [PMID: 22851042 DOI: 10.4414/smw.2012.13653] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
"Epimmunome", a term introduced recently by Swamy and colleagues, describes all molecules and pathways used by epithelial cells (ECs) to instruct immune cells. Today, we know that ECs are among the first sites within the human body to be exposed to pathogens (such as influenza viruses) and that the release of chemokine and cytokines by ECs is influenced by inhaled agents. The role of the ECs as a switchboard to initiate and regulate immune responses is altered through air pollutant exposure, such as ozone, tobacco smoke and diesel exhaust emissions. The details of the interplay between ECs and immune cells are not yet fully understood and need to be investigated further. Co-culture models, cell specific genetically-modified mice and the analysis of human biopsies provide great tools to gain knowledge about potential mechanisms. Increasing our understanding about the role of ECs in respiratory immunity may yield novel therapeutic targets to modulate downstream diseases.
Collapse
Affiliation(s)
- Loretta Müller
- Center for Environmental Medicine, Asthma and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7310, USA.
| | | |
Collapse
|
25
|
Pazos MA, Kraus TA, Muñoz-Fontela C, Moran TM. Estrogen mediates innate and adaptive immune alterations to influenza infection in pregnant mice. PLoS One 2012; 7:e40502. [PMID: 22792357 PMCID: PMC3390370 DOI: 10.1371/journal.pone.0040502] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 06/12/2012] [Indexed: 11/19/2022] Open
Abstract
Pregnancy is a leading risk factor for severe complications during an influenza virus infection. Women infected during their second and third trimesters are at increased risk for severe cardiopulmonary complications, premature delivery, and death. Here, we establish a murine model of aerosolized influenza infection during pregnancy. We find significantly altered innate antiviral responses in pregnant mice, including decreased levels of IFN-β, IL-1α, and IFN-γ at early time points of infection. We also find reduced cytotoxic T cell activity and delayed viral clearance. We further demonstrate that pregnancy levels of the estrogen 17-β-estradiol are able to induce key anti-inflammatory phenotypes in immune responses to the virus independently of other hormones or pregnancy-related stressors. We conclude that elevated estrogen levels result in an attenuated anti-viral immune response, and that pregnancy-associated morbidities occur in the context of this anti-inflammatory phenotype.
Collapse
Affiliation(s)
- Michael A. Pazos
- Department of Microbiology, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Thomas A. Kraus
- Department of Microbiology, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - César Muñoz-Fontela
- Heinrich-Pette-Institut, Leibniz-Institut für Experimentelle Virologie, Hamburg, Germany
| | - Thomas M. Moran
- Department of Microbiology, Mount Sinai School of Medicine, New York, New York, United States of America
| |
Collapse
|
26
|
Hermesh T, Moran TM, Jain D, López CB. Granulocyte colony-stimulating factor protects mice during respiratory virus infections. PLoS One 2012; 7:e37334. [PMID: 22615983 PMCID: PMC3353936 DOI: 10.1371/journal.pone.0037334] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Accepted: 04/19/2012] [Indexed: 01/13/2023] Open
Abstract
A burst in the production of pro-inflammatory molecules characterizes the beginning of the host response to infection. Cytokines, chemokines, and growth factors work in concert to control pathogen replication and activate innate and adaptive immune responses. Granulocyte colony-stimulating factor (G-CSF) mobilizes and activates hematopoietic cells from the bone marrow, and it has been shown to mediate the generation of effective immunity against bacterial and fungal infections. G-CSF is produced at high levels in the lungs during infection with influenza and parainfluenza viruses, but its role during these infections is unknown. Here we show that during infection of mice with a non-lethal dose of influenza or Sendai virus, G-CSF promotes the accumulation of activated Ly6G+ granulocytes that control the extent of the lung pro-inflammatory response. Remarkably, these G-CSF-mediated effects facilitate viral clearance and sustain mouse survival.
Collapse
Affiliation(s)
- Tamar Hermesh
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Thomas M. Moran
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Deepika Jain
- Department of Pathobiology School of Veterinary Medicine and Institute for Immunology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Carolina B. López
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Pathobiology School of Veterinary Medicine and Institute for Immunology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
27
|
Reuter S, Dehzad N, Martin H, Böhm L, Becker M, Buhl R, Stassen M, Taube C. TLR3 but not TLR7/8 ligand induces allergic sensitization to inhaled allergen. THE JOURNAL OF IMMUNOLOGY 2012; 188:5123-31. [PMID: 22491246 DOI: 10.4049/jimmunol.1101618] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Epidemiological studies suggest that viral infections during childhood are a risk factor for the development of asthma. However, the role of virus-specific pattern recognition receptors in this process is not well defined. In the current study, we compare the effects of the inhaled viral TLR ligands polyinosinic-polycytidylic acid (TLR3) and resiquimod (TLR7/8) on sensitization to a model allergen (OVA) in a murine model. Both compounds enhance the migration, activation, and Ag-processing of myeloid dendritic cells from the lung to the draining lymph nodes comparable to the effects of LPS. Application of polyinosinic-polycytidylic acid [poly(I:C)] or LPS induces production of allergen-specific IgE and IgG1, whereas resiquimod (R848) had no effect. In addition, rechallenge of mice with OVA resulted in airway inflammation and mucus production in animals that received either poly(I:C) or LPS but not after application of R848. In summary, these results show that activation of TLR3 in combination with inhaled allergen results in induction of dendritic cell activation and migration similar to the effects of LPS. This leads to the development of allergic airway disease after allergen rechallenge, whereas mice treated with R848 did not develop allergic airway disease. These findings give further insight into the effects of stimulation of different TLRs on the development of asthma.
Collapse
Affiliation(s)
- Sebastian Reuter
- III Department of Medicine, University Hospital Mainz, Mainz, Germany
| | | | | | | | | | | | | | | |
Collapse
|
28
|
de Geus ED, Jansen CA, Vervelde L. Uptake of Particulate Antigens in a Nonmammalian Lung: Phenotypic and Functional Characterization of Avian Respiratory Phagocytes Using Bacterial or Viral Antigens. THE JOURNAL OF IMMUNOLOGY 2012; 188:4516-26. [DOI: 10.4049/jimmunol.1200092] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
29
|
Lambrecht BN, Hammad H. Lung dendritic cells in respiratory viral infection and asthma: from protection to immunopathology. Annu Rev Immunol 2012; 30:243-70. [PMID: 22224777 DOI: 10.1146/annurev-immunol-020711-075021] [Citation(s) in RCA: 232] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Lung dendritic cells (DCs) bridge innate and adaptive immunity, and depending on context, they also induce a Th1, Th2, or Th17 response to optimally clear infectious threats. Conversely, lung DCs can also mount maladaptive Th2 immune responses to harmless allergens and, in this way, contribute to immunopathology. It is now clear that the various aspects of DC biology can be understood only if we take into account the functional specializations of different DC subsets that are present in the lung in homeostasis or are attracted to the lung as part of the inflammatory response to inhaled noxious stimuli. Lung DCs are heavily influenced by the nearby epithelial cells, and a model is emerging whereby direct communication between DCs and epithelial cells determines the outcome of the pulmonary immune response. Here, we have approached DC biology from the perspective of viral infection and allergy to illustrate these emerging concepts.
Collapse
Affiliation(s)
- Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, Department of Molecular Biomedical Research, VIB, 9052 Ghent, Belgium.
| | | |
Collapse
|
30
|
Moltedo B, Li W, Yount JS, Moran TM. Unique type I interferon responses determine the functional fate of migratory lung dendritic cells during influenza virus infection. PLoS Pathog 2011; 7:e1002345. [PMID: 22072965 PMCID: PMC3207893 DOI: 10.1371/journal.ppat.1002345] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Accepted: 09/15/2011] [Indexed: 12/24/2022] Open
Abstract
Migratory lung dendritic cells (DCs) transport viral antigen from the lungs to the draining mediastinal lymph nodes (MLNs) during influenza virus infection to initiate the adaptive immune response. Two major migratory DC subsets, CD103+ DCs and CD11bhigh DCs participate in this function and it is not clear if these antigen presenting cell (APC) populations become directly infected and if so whether their activity is influenced by the infection. In these experiments we show that both subpopulations can become infected and migrate to the draining MLN but a difference in their response to type I interferon (I-IFN) signaling dictates the capacity of the virus to replicate. CD103+ DCs allow the virus to replicate to significantly higher levels than do the CD11bhigh DCs, and they release infectious virus in the MLNs and when cultured ex-vivo. Virus replication in CD11bhigh DCs is inhibited by I-IFNs, since ablation of the I-IFN receptor (IFNAR) signaling permits virus to replicate vigorously and productively in this subset. Interestingly, CD103+ DCs are less sensitive to I-IFNs upregulating interferon-induced genes to a lesser extent than CD11bhigh DCs. The attenuated IFNAR signaling by CD103+ DCs correlates with their described superior antigen presentation capacity for naïve CD8+ T cells when compared to CD11bhigh DCs. Indeed ablation of IFNAR signaling equalizes the competency of the antigen presenting function for the two subpopulations. Thus, antigen presentation by lung DCs is proportional to virus replication and this is tightly constrained by I-IFN. The “interferon-resistant” CD103+ DCs may have evolved to ensure the presentation of viral antigens to T cells in I-IFN rich environments. Conversely, this trait may be exploitable by viral pathogens as a mechanism for systemic dissemination. Migratory lung dendritic cells (DCs) control the initiation of the adaptive immune responses to influenza virus by expanding virus-specific T cells in draining lymph nodes (MLNs) that will subsequently clear the pathogen from the respiratory tract. Here we demonstrate that both subsets of lung DCs, CD103+ DCs and CD11bhigh DCs become infected by influenza virus in vivo and migrate to the MLNs, but only CD103+ DCs support productive virus replication. Enhanced virus replication in CD103+ DCs compared to CD11bhigh DCs was responsible for their superior antigen presentation efficacy for naïve CD8+ T cells and originated from a difference in sensitivity of the two DC populations to type I interferon (I-IFN). These data show that in contrast to most other immune cell types, DCs can become productively infected with influenza virus and I-IFN operates as a master regulator controlling which DC subset will present antigen during a viral infection. A deeper understanding of basic innate and adaptive immune response mechanisms regulated by I-FN may lead to the development of cutting edge therapies and improve vaccine efficacy against influenza and other viruses.
Collapse
Affiliation(s)
- Bruno Moltedo
- Department of Microbiology, Mount Sinai School of Medicine, New York, New York, United States of America
- Immunology Institute, Mount Sinai School of Medicine, New York, New York, United States of America
- * E-mail: (TMM); (BM)
| | - Wenjing Li
- Department of Microbiology, Mount Sinai School of Medicine, New York, New York, United States of America
- Immunology Institute, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Jacob S. Yount
- Department of Microbiology, Mount Sinai School of Medicine, New York, New York, United States of America
- Immunology Institute, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Thomas M. Moran
- Department of Microbiology, Mount Sinai School of Medicine, New York, New York, United States of America
- Immunology Institute, Mount Sinai School of Medicine, New York, New York, United States of America
- * E-mail: (TMM); (BM)
| |
Collapse
|
31
|
Fear VS, Burchell JT, Lai SP, Wikstrom ME, Blank F, von Garnier C, Turner DJ, Sly PD, Holt PG, Strickland DS, Stumbles PA. Restricted Aeroallergen Access to Airway Mucosal Dendritic Cells In Vivo Limits Allergen-Specific CD4+ T Cell Proliferation during the Induction of Inhalation Tolerance. THE JOURNAL OF IMMUNOLOGY 2011; 187:4561-70. [DOI: 10.4049/jimmunol.1004189] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
32
|
Pneumocystis infection in an immunocompetent host can promote collateral sensitization to respiratory antigens. Infect Immun 2011; 79:1905-14. [PMID: 21343358 DOI: 10.1128/iai.01273-10] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Infection with the opportunistic fungal pathogen Pneumocystis is assumed to pass without persistent pathology in immunocompetent hosts. However, when immunocompetent BALB/c mice were inoculated with Pneumocystis, a vigorous Th2-like pulmonary inflammation ensued and peaked at 14 days postinfection. This coincided with a 10-fold increase in the number of antigen-presenting cells (APCs) in the lung, and these cells were capable of presenting antigen in vitro, as well as greater uptake of antigen in vivo. When mice were presented with exogenous antigen at the 14-day time point of the infection, they developed respiratory sensitization to that antigen, in the form of increased airway hyperresponsiveness upon a later challenge, whereas mice not infected but presented with antigen did not. Like other forms of collateral sensitization, this response was dependent on interleukin-4 receptor signaling. This ability to facilitate sensitization to exogenous antigen has been previously reported for other infectious disease agents; however, Pneumocystis appears to be uniquely capable in this respect, as a single intranasal dose without added adjuvant, when it was administered at the appropriate time, was sufficient to initiate sensitization. Pneumocystis infection probably occurs in most humans during the first few years of life, and in the vast majority of cases, it fails to cause any overt direct pathology. However, as we show here, Pneumocystis can be an agent of comorbidity at this time by facilitating respiratory sensitization that may relate to the later development or exacerbation of obstructive airway disease.
Collapse
|
33
|
Pedroza-Gonzalez A, Xu K, Wu TC, Aspord C, Tindle S, Marches F, Gallegos M, Burton EC, Savino D, Hori T, Tanaka Y, Zurawski S, Zurawski G, Bover L, Liu YJ, Banchereau J, Palucka AK. Thymic stromal lymphopoietin fosters human breast tumor growth by promoting type 2 inflammation. ACTA ACUST UNITED AC 2011; 208:479-90. [PMID: 21339324 PMCID: PMC3058586 DOI: 10.1084/jem.20102131] [Citation(s) in RCA: 187] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The human breast tumor microenvironment can display features of T helper type 2 (Th2) inflammation, and Th2 inflammation can promote tumor development. However, the molecular and cellular mechanisms contributing to Th2 inflammation in breast tumors remain unclear. Here, we show that human breast cancer cells produce thymic stromal lymphopoietin (TSLP). Breast tumor supernatants, in a TSLP-dependent manner, induce expression of OX40L on dendritic cells (DCs). OX40L(+) DCs are found in primary breast tumor infiltrates. OX40L(+) DCs drive development of inflammatory Th2 cells producing interleukin-13 and tumor necrosis factor in vitro. Antibodies neutralizing TSLP or OX40L inhibit breast tumor growth and interleukin-13 production in a xenograft model. Thus, breast cancer cell-derived TSLP contributes to the inflammatory Th2 microenvironment conducive to breast tumor development by inducing OX40L expression on DCs.
Collapse
|
34
|
Abstract
T cells can reject established tumours when adoptively transferred into patients, thereby demonstrating the power of the immune system for cancer therapy. However, it has proven difficult to maintain adoptively transferred T cells in the long term. Vaccines have the potential to induce tumour-specific effector and memory T cells. However, clinical efficacy of current vaccines is limited, possibly because tumours skew the immune system by means of myeloid-derived suppressor cells, inflammatory type 2 T cells and regulatory T cells (Tregs), all of which prevent the generation of effector cells. To improve the clinical efficacy of cancer vaccines in patients with metastatic disease, we need to design novel and improved strategies that can boost adaptive immunity to cancer, help overcome Tregs and allow the breakdown of the immunosuppressive tumour microenvironment. This can be achieved by exploiting the fast increasing knowledge about the dendritic cell (DC) system, including the existence of distinct DC subsets that respond differentially to distinct activation signals, (functional plasticity), both contributing to the generation of unique adaptive immune responses. We foresee that these novel cancer vaccines will be used as monotherapy in patients with resected disease and in combination with drugs targeting regulatory/suppressor pathways in patients with metastatic disease.
Collapse
Affiliation(s)
- K Palucka
- Baylor Institute for Immunology Research, Baylor University Medical Center, Dallas, TX, USA.
| | | | | | | |
Collapse
|
35
|
Plantinga M, Hammad H, Lambrecht BN. Origin and functional specializations of DC subsets in the lung. Eur J Immunol 2010; 40:2112-8. [PMID: 20853496 DOI: 10.1002/eji.201040562] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Lung DC bridge innate and adaptive immunity, and depending on the context, induce Th1, Th2 or Th17 response, to optimally clear infections. Conversely, lung DC can also prevent overt and harmful immune responses to harmless inhaled antigens via induction of Treg cells or via induction of neutralizing mucosal IgA antibodies. Here, we propose that these functions are not the result of a single population of DC, and instead, subsets of DC perform specialized functions.
Collapse
Affiliation(s)
- Maud Plantinga
- Laboratory of Immunoregulation and Mucosal Immunology, Department of Respiratory Diseases, Ghent University, Belgium
| | | | | |
Collapse
|
36
|
Abstract
This Viewpoint series provides authoritative and detailed outlines of exciting areas of DC research. Some of the subjects that frequently come up include development of DC; distribution of DC in lymphoid and non-lymphoid tissues such as skin, intestine and lung; different forms or subsets of DC; and the role of DC in initiating tolerance and immunity. In this Preface, I will introduce the Viewpoints and consider some future challenges as well as the medical relevance of DC research.
Collapse
|
37
|
Hermesh T, Moltedo B, López CB, Moran TM. Buying time-the immune system determinants of the incubation period to respiratory viruses. Viruses 2010; 2:2541-58. [PMID: 21994630 PMCID: PMC3185581 DOI: 10.3390/v2112541] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Revised: 11/01/2010] [Accepted: 11/02/2010] [Indexed: 12/20/2022] Open
Abstract
Respiratory viruses cause disease in humans characterized by an abrupt onset of symptoms. Studies in humans and animal models have shown that symptoms are not immediate and appear days or even weeks after infection. Since the initial symptoms are a manifestation of virus recognition by elements of the innate immune response, early virus replication must go largely undetected. The interval between infection and the emergence of symptoms is called the incubation period and is widely used as a clinical score. While incubation periods have been described for many virus infections the underlying mechanism for this asymptomatic phase has not been comprehensively documented. Here we review studies of the interaction between human pathogenic respiratory RNA viruses and the host with a particular emphasis on the mechanisms used by viruses to inhibit immunity. We discuss the concept of the "stealth phase", defined as the time between infection and the earliest detectable inflammatory response. We propose that the "stealth phase" phenomenon is primarily responsible for the suppression of symptoms during the incubation period and results from viral antagonism that inhibits major pathways of the innate immune system allowing an extended time of unhindered virus replication.
Collapse
Affiliation(s)
- Tamar Hermesh
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029, USA; E-Mails: (T.H.); (B.M.); (C.B.L.)
| | - Bruno Moltedo
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029, USA; E-Mails: (T.H.); (B.M.); (C.B.L.)
| | - Carolina B. López
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029, USA; E-Mails: (T.H.); (B.M.); (C.B.L.)
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Thomas M. Moran
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029, USA; E-Mails: (T.H.); (B.M.); (C.B.L.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-212-241-7963; Fax: +1-212-987-3653
| |
Collapse
|
38
|
Functional dichotomy between NKG2D and CD28-mediated co-stimulation in human CD8+ T cells. PLoS One 2010; 5. [PMID: 20844584 PMCID: PMC2936560 DOI: 10.1371/journal.pone.0012635] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Accepted: 08/16/2010] [Indexed: 11/19/2022] Open
Abstract
Both CD28 and NKG2D can function as co-stimulatory receptors in human CD8+ T cells. However, their independent functional contributions in distinct CD8+ T cell subsets are not well understood. In this study, CD8+ T cells in human peripheral blood- and lung-derived lymphocytes were analyzed for CD28 and NKG2D expression and function. We found a higher level of CD28 expression in PBMC-derived naïve (CD45RA+CD27+) and memory (CD45RA−CD27+) CD8+ T cells (CD28Hi), while its expression was significantly lower in effector (CD45RA+CD27−) CD8+ T cells (CD28Lo). Irrespective of the differences in the CD28 levels, NKG2D expression was comparable in all three CD8+ T cell subsets. CD28 and NKG2D expressions followed similar patterns in human lung-resident GILGFVFTL/HLA-A2-pentamer positive CD8+ T cells. Co-stimulation of CD28Lo effector T cells via NKG2D significantly increased IFN-γ and TNF-α levels. On the contrary, irrespective of its comparable levels, NKG2D-mediated co-stimulation failed to augment IFN-γ and TNF-α production in CD28Hi naïve/memory T cells. Additionally, CD28-mediated co-stimulation was obligatory for IL-2 generation and thereby its production was limited only to the CD28Hi naïve/memory subsets. MICA, a ligand for NKG2D was abundantly expressed in the tracheal epithelial cells, validating the use of NKG2D as the major co-stimulatory receptor by tissue-resident CD8+ effector T cells. Based on these findings, we conclude that NKG2D may provide an expanded level of co-stimulation to tissue-residing effector CD8+ T cells. Thus, incorporation of co-stimulation via NKG2D in addition to CD28 is essential to activate tumor or tissue-infiltrating effector CD8+ T cells. However, boosting a recall immune response via memory CD8+ T cells or vaccination to stimulate naïve CD8+ T cells would require CD28-mediated co-stimulation.
Collapse
|
39
|
Mount AM, Masson F, Kupresanin F, Smith CM, May JS, van Rooijen N, Stevenson PG, Belz GT. Interference with dendritic cell populations limits early antigen presentation in chronic γ-herpesvirus-68 infection. THE JOURNAL OF IMMUNOLOGY 2010; 185:3669-76. [PMID: 20720208 DOI: 10.4049/jimmunol.1001079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A critical factor influencing the ability of the host to mount a robust immune response against a virus depends on the rapid recruitment of dendritic cells (DCs) presenting Ags. From the outset, this step sets the tempo for subsequent activation of virus-specific T cells. Despite this, how induction of the immune response might be modified by pathogens with the capacity to establish persistence is unclear. In this study, we have characterized the in vivo influence of murine gamma-herpesvirus K3-mediated interference with MHC class I in DCs that drive the initial adaptive immune response. We observed that gamma-herpesvirus could interfere with the very earliest phase of Ag presentation through K3 by directly targeting migratory and lymph node-resident DCs. These results show that a pathogen with the capacity to interfere with early Ag presentation can establish suboptimal conditions for rapid induction of the adaptive immune response and thus favor establishment of viral persistence.
Collapse
Affiliation(s)
- Adele M Mount
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Drutman SB, Trombetta ES. Dendritic cells continue to capture and present antigens after maturation in vivo. THE JOURNAL OF IMMUNOLOGY 2010; 185:2140-6. [PMID: 20644175 DOI: 10.4049/jimmunol.1000642] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Dendritic cell (DC) maturation is critical for the regulation of T cell responses. The downregulation of endocytosis on maturation is considered a key adaptation that dissociates prior Ag capture by DCs from subsequent T cell engagement. To study the dynamics of Ag capture and presentation in situ, we studied the capacity for Ag uptake by DCs matured in their natural tissue environment. We found that after maturation in vivo, mouse DCs retained a robust capacity to capture soluble Ags. Furthermore, Ags internalized by mature DCs were efficiently presented on MHC class II and cross-presented on MHC class I. These results suggest that under inflammatory conditions, mature DCs may contribute to T cell stimulation without exclusively relying on prior exposure to Ags as immature DC precursors.
Collapse
Affiliation(s)
- Scott B Drutman
- Cancer Institute, New York University School of Medicine, New York, NY 10016, USA
| | | |
Collapse
|
41
|
Abstract
Cancer vaccines have undergone a renaissance because of recent clinical trials showing promising immunologic data and some clinical benefit to patients. Current trials exploiting dendritic cells (DCs) as vaccines have shown durable tumor regressions in a fraction of patients. Clinical efficacy of current vaccines is hampered by myeloid-derived suppressor cells, inflammatory type 2 T cells, and regulatory T cells, all of which prevent the generation of effector cells. To improve the clinical efficacy of DC vaccines, we need to design novel and improved strategies that can boost adaptive immunity to cancer, help overcome regulatory T cells and allow the breakdown of the immunosuppressive tumor microenvironment. This can be achieved by exploiting the fast increasing knowledge about the DC system, including the existence of distinct DC subsets. Critical to the design of better vaccines is the concept of distinct DC subsets and distinct DC activation pathways, all contributing to the generation of unique adaptive immune responses. Such novel DC vaccines will be used as monotherapy in patients with resected disease and in combination with antibodies and/or drugs targeting suppressor pathways and modulation of the tumor environment in patients with metastatic disease.
Collapse
Affiliation(s)
- Karolina Palucka
- Baylor Institute for Immunology Research, Dallas, TX 75204, USA.
| | | | | | | | | |
Collapse
|
42
|
Nörder M, Becker PD, Drexler I, Link C, Erfle V, Guzmán CA. Modified vaccinia virus Ankara exerts potent immune modulatory activities in a murine model. PLoS One 2010; 5:e11400. [PMID: 20628596 PMCID: PMC2900180 DOI: 10.1371/journal.pone.0011400] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Accepted: 05/31/2010] [Indexed: 11/18/2022] Open
Abstract
Background Modified vaccinia virus Ankara (MVA), a highly attenuated strain of vaccinia virus, has been used as vaccine delivery vector in preclinical and clinical studies against infectious diseases and malignancies. Here, we investigated whether an MVA which does not encode any antigen (Ag) could be exploited as adjuvant per se. Methodology/Principal Findings We showed that dendritic cells infected in vitro with non-recombinant (nr) MVA expressed maturation and activation markers and were able to efficiently present exogenously pulsed Ag to T cells. In contrast to the dominant T helper (Th) 1 biased responses elicited against Ags produced by recombinant MVA vectors, the use of nrMVA as adjuvant for the co-administered soluble Ags resulted in a long lasting mixed Th1/Th2 responses. Conclusions/Significance These findings open new ways to potentiate and modulate the immune responses to vaccine Ags depending on whether they are co-administered with MVA or encoded by recombinant viruses.
Collapse
Affiliation(s)
- Miriam Nörder
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Pablo D. Becker
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ingo Drexler
- Institute of Virology, Technische Universität München and Helmholtz Centre Munich, Munich, Germany
| | - Claudia Link
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Volker Erfle
- Institute of Virology, Technische Universität München and Helmholtz Centre Munich, Munich, Germany
- Clinical Cooperation Group Antigen Specific Immunomodulation, Technische Universität München and Helmholtz Centre Munich, Munich, Germany
| | - Carlos A. Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- * E-mail:
| |
Collapse
|
43
|
Maggina P, Christodoulou I, Papaevangelou V, Tsolia M, Papadopoulos NG. Dendritic cells in viral bronchiolitis. Expert Rev Clin Immunol 2010; 5:271-82. [PMID: 20477005 DOI: 10.1586/eci.09.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Dendritic cells (DCs) are major antigen-presenting cells that constitute a link between innate and adaptive immune responses, and are critical in the processes of control and elimination of viral infections. On the other hand, there is a large body of data strongly implicating respiratory viruses in morbidity during infancy through the induction of lower respiratory tract infections, such as bronchiolitis, and later on in childhood and adult life, mainly due to their association with asthma exacerbations. Little is known, however, about the precise role of DCs in human respiratory tract infections. This review focuses on current data, both from in vivo and in vitro studies, that highlight the interplay between DCs and the viral causes of bronchiolitis.
Collapse
Affiliation(s)
- Paraskevi Maggina
- Allergy Research Centre, 2nd Paediatric Clinic, Medical School, University of Athens, 41 Fidippidou Street, Goudi, 11527 Athens, Greece.
| | | | | | | | | |
Collapse
|
44
|
Aronica MA, Vogel N. Pathogens and immunologic memory in asthma: what have we learned? Expert Rev Clin Immunol 2010; 1:589-601. [PMID: 20477600 DOI: 10.1586/1744666x.1.4.589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Animal models and clinical studies of asthma have generated important insights into the first effector phase leading to the development of allergic airway disease and bronchial hyper-reactivity. In contrast, mechanisms related to asthma chronicity or persistence are less well understood. The CD4(+) T-helper 2 lymphocytes are known initiators of the inflammatory response associated with asthma. There is now increasing evidence that memory T-cells, sensitized against allergenic, occupational or viral antigens, are also involved in the persistence of asthma. Additionally, the role of pathogens in asthma has been linked to both the initial susceptibility to and flares of this disease. This review will discuss the potential links between infection and asthma, the role of the memory T-cells in asthma, and the potential mechanisms by which these factors interact to lead to the development and/or persistence of asthma.
Collapse
Affiliation(s)
- Mark A Aronica
- Cleveland Clinic Foundation, Department of Pulmonary, Allergy and Critical Care Medicine, Cleveland, OH 44195, USA.
| | | |
Collapse
|
45
|
Jin GB, Moore AJ, Head JL, Neumiller JJ, Lawrence BP. Aryl hydrocarbon receptor activation reduces dendritic cell function during influenza virus infection. Toxicol Sci 2010; 116:514-22. [PMID: 20498003 DOI: 10.1093/toxsci/kfq153] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
It has long been known that activation of the aryl hydrocarbon receptor (AhR) by ligands such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) suppresses T cell-dependent immune responses; however, the underlying cellular targets and mechanism remain unclear. We have previously shown that AhR activation by TCDD reduces the proliferation and differentiation of influenza virus-specific CD8(+) T cells through an indirect mechanism; suggesting that accessory cells are critical AhR targets during infection. Respiratory dendritic cells (DCs) capture antigen, migrate to lymph nodes, and play a key role in activating naive CD8(+) T cells during respiratory virus infection. Herein, we report an examination of how AhR activation alters DCs in the lung and affects their trafficking to and function in the mediastinal lymph nodes (MLN) during infection with influenza virus. We show that AhR activation impairs lung DC migration and reduces the ability of DCs isolated from the MLN to activate naive CD8(+) T cells. Using novel AhR mutant mice, in which the AhR protein lacks its DNA-binding domain, we show that the suppressive effects of TCDD require that the activated AhR complex binds to DNA. These new findings suggest that AhR activation by chemicals from our environment impacts DC function to stimulate naive CD8(+) T cells and that immunoregulatory genes within DCs are critical targets of AhR. Moreover, our results reinforce the idea that environmental signals and AhR ligands may contribute to differential susceptibilities and responses to respiratory infection.
Collapse
Affiliation(s)
- Guang-Bi Jin
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | | | | | | | | |
Collapse
|
46
|
Qiao L, Phipps-Yonas H, Hartmann B, Moran TM, Sealfon SC, Hayot F. Immune response modeling of interferon beta-pretreated influenza virus-infected human dendritic cells. Biophys J 2010; 98:505-14. [PMID: 20159146 DOI: 10.1016/j.bpj.2009.10.049] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2009] [Revised: 09/18/2009] [Accepted: 10/30/2009] [Indexed: 12/20/2022] Open
Abstract
The pretreatment of human dendritic cells with interferon-beta enhances their immune response to influenza virus infection. We measured the expression levels of several key players in that response over a period of 13 h both during pretreatment and after viral infection. Their activation profiles reflect the presence of both negative and positive feedback loops in interferon induction and interferon signaling pathway. Based on these measurements, we have developed a comprehensive computational model of cellular immune response that elucidates its mechanism and its dynamics in interferon-pretreated dendritic cells, and provides insights into the effects of duration and strength of pretreatment.
Collapse
Affiliation(s)
- Liang Qiao
- Department of Neurology, Mount Sinai School of Medicine, New York, New York, USA
| | | | | | | | | | | |
Collapse
|
47
|
Schmolke M, García-Sastre A. Evasion of innate and adaptive immune responses by influenza A virus. Cell Microbiol 2010; 12:873-80. [PMID: 20482552 DOI: 10.1111/j.1462-5822.2010.01475.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Host organisms have developed sophisticated antiviral responses in order to defeat emerging influenza A viruses (IAVs). At the same time IAVs have evolved immune evasion strategies. The immune system of mammals provides several lines of defence to neutralize invading pathogens or limit their replication. Here, we summarize the mammalian innate and adaptive immune mechanisms involved in host defence against viral infection and review strategies by which IAVs avoid, circumvent or subvert these mechanisms. We highlight well-characterized, as well as recently described features of this intriguing virus-host molecular battle.
Collapse
Affiliation(s)
- Mirco Schmolke
- Department of Microbiology, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA
| | | |
Collapse
|
48
|
Abstract
The Banting Medal for Scientific Achievement Award is the American Diabetes Association's highest scientific award and honors an individual who has made significant, long-term contributions to the understanding of diabetes, its treatment, and/or prevention. The award is named after Nobel Prize winner Sir Frederick Banting, who codiscovered insulin treatment for diabetes. Dr. Eisenbarth received the American Diabetes Association's Banting Medal for Scientific Achievement at the Association's 69th Scientific Sessions, June 5–9, 2009, in New Orleans, Louisiana. He presented the Banting Lecture, An Unfinished Journey—Type 1 Diabetes—Molecular Pathogenesis to Prevention , on Sunday, June 7, 2009.
Collapse
Affiliation(s)
- George S Eisenbarth
- Barbara Davis Center for Childhood Diabetes, University of Colorado Health Sciences Center, Aurora, Colorado, USA.
| |
Collapse
|
49
|
Wikstrom ME, Batanero E, Judd SR, Wiqvist K, Holt PG, Stumbles PA. Lung homing T-cell generation is dependent on strength and timing of antigen delivery to lymph nodes. Immunol Cell Biol 2010; 88:658-66. [PMID: 20195280 DOI: 10.1038/icb.2010.18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Inhaled allergens are known for their immediate and ongoing effects in the respiratory tract (RT). In this report, we track inhaled antigen in normal mice for 7 days and find that while it is cleared from the airways, inhaled antigen persists in peripheral lung tissue and the draining lymph nodes (DLNs). The persistence of antigen led to ongoing presentation in the lymph nodes, but not the lungs, that decreased with time in direct proportion with the frequency of antigen-bearing RT dendritic cells (DCs). There was evidence of functional changes among the antigen-bearing DCs in the lymph nodes, as the expression of CD40, CD80 and CD86 were modulated over the course of 7 days. At the same time, there was a decrease in both CD4(+) T-cell proliferation in lymph nodes and the generation of recirculating CD4(+) T cells. However, early presentation of lower doses of inhaled antigen also resulted in a decrease in CD4(+) T-cell proliferation and recirculation. Thus, T-cell recirculation depends on the strength of stimulus in the DLNs and is produced by a combination of the dose of antigen delivered to the RT, DC migration and co-stimulatory molecule expression. These results provide an important insight into the fate of inhaled antigen in vivo and the influence of persistent antigen presentation on T-cell activation in the lymph nodes.
Collapse
Affiliation(s)
- Matthew E Wikstrom
- Telethon Institute for Child Health Research and Centre for Child Health Research, School of Paediatrics and Child Health, University of Western Australia, Perth, Western Australia, Australia
| | | | | | | | | | | |
Collapse
|
50
|
Huvenne W, Callebaut I, Plantinga M, Vanoirbeek JAJ, Krysko O, Bullens DMA, Gevaert P, Van Cauwenberge P, Lambrecht BN, Ceuppens JL, Bachert C, Hellings PW. Staphylococcus aureus enterotoxin B facilitates allergic sensitization in experimental asthma. Clin Exp Allergy 2010; 40:1079-90. [PMID: 20214664 DOI: 10.1111/j.1365-2222.2010.03464.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Staphylococcus aureus Enterotoxin B (SEB) has immunomodulatory effects in allergic airway disease. The potential contribution of SEB to the sensitization process to allergens remains obscure. OBJECTIVE In order to study the effects of staphylococcal-derived toxins on the sensitization to ovalbumin (OVA) and induction of allergic airway inflammation, we have combined the nasal application of OVA with different toxins. METHODS Nasal applications of OVA and saline, SEA, SEB, toxic shock syndrome toxin (TSST)-1, protein A or lipopolysaccharide (LPS) were performed on alternate days from day 0 till 12. On day 14, mice were killed for the evaluation of OVA-specific IgE, cytokine production by mediastinal lymph node (MLN) cells and bronchial hyperreactivity (BHR) to inhaled metacholine. The effect of SEB on dendritic cell (DC) migration and maturation, and on T cell proliferation was evaluated. RESULTS Concomitant endonasal application of OVA and SEB resulted in OVA-specific IgE production, whereas this was not found with SEA, TSST-1, protein A, LPS or OVA alone. Increased DC maturation and migration to the draining lymph nodes were observed in OVA/SEB mice, as well as an increased T cell proliferation. Bronchial inflammation with an influx of eosinophils and lymphocytes was demonstrated in OVA/SEB mice, together with hyperresponsiveness and the production of IL-4, IL-5, IL-10 and IL-13 by MLN stimulated with OVA. CONCLUSIONS Our data demonstrate that SEB facilitates sensitization to OVA and consecutive bronchial inflammation with features of allergic asthma. This is likely due to augmentation of DC migration and maturation, as well as the allergen-specific T cell proliferation upon concomitant OVA and SEB application.
Collapse
Affiliation(s)
- W Huvenne
- Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University Hospital, Ghent, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|