1
|
Hargrave KE, Worrell JC, Pirillo C, Brennan E, Masdefiol Garriga A, Gray JI, Purnell T, Roberts EW, MacLeod MKL. Lung influenza virus-specific memory CD4 T cell location and optimal cytokine production are dependent on interactions with lung antigen-presenting cells. Mucosal Immunol 2024; 17:843-857. [PMID: 38851589 PMCID: PMC11464401 DOI: 10.1016/j.mucimm.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 05/29/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
Influenza A virus (IAV) infection leads to the formation of mucosal memory CD4 T cells that can protect the host. An in-depth understanding of the signals that shape memory cell development is required for more effective vaccine design. We have examined the formation of memory CD4 T cells in the lung following IAV infection of mice, characterizing changes to the lung landscape and immune cell composition. IAV-specific CD4 T cells were found throughout the lung at both primary and memory time points. These cells were found near lung airways and in close contact with a range of immune cells including macrophages, dendritic cells, and B cells. Interactions between lung IAV-specific CD4 T cells and major histocompatibility complex (MHC)II+ cells during the primary immune response were important in shaping the subsequent memory pool. Treatment with an anti-MHCII blocking antibody increased the proportion of memory CD4 T cells found in lung airways but reduced interferon-γ expression by IAV-specific immunodominant memory CD4 T cells. The immunodominant CD4 T cells expressed higher levels of programmed death ligand 1 (PD1) than other IAV-specific CD4 T cells and PD1+ memory CD4 T cells were located further away from MHCII+ cells than their PD1-low counterparts. This distinction in location was lost in mice treated with anti-MHCII antibodies. These data suggest that sustained antigen presentation in the lung impacts the formation of memory CD4 T cells by regulating their cytokine production and location.
Collapse
Affiliation(s)
- Kerrie E Hargrave
- Centre for Immunobiology, School of Infection and Immunity, University of Glasgow, UK
| | - Julie C Worrell
- Centre for Immunobiology, School of Infection and Immunity, University of Glasgow, UK
| | | | - Euan Brennan
- Centre for Immunobiology, School of Infection and Immunity, University of Glasgow, UK
| | | | - Joshua I Gray
- Centre for Immunobiology, School of Infection and Immunity, University of Glasgow, UK
| | - Thomas Purnell
- Centre for Immunobiology, School of Infection and Immunity, University of Glasgow, UK
| | | | - Megan K L MacLeod
- Centre for Immunobiology, School of Infection and Immunity, University of Glasgow, UK.
| |
Collapse
|
2
|
Finn CM, McKinstry KK. Ex Pluribus Unum: The CD4 T Cell Response against Influenza A Virus. Cells 2024; 13:639. [PMID: 38607077 PMCID: PMC11012043 DOI: 10.3390/cells13070639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024] Open
Abstract
Current Influenza A virus (IAV) vaccines, which primarily aim to generate neutralizing antibodies against the major surface proteins of specific IAV strains predicted to circulate during the annual 'flu' season, are suboptimal and are characterized by relatively low annual vaccine efficacy. One approach to improve protection is for vaccines to also target the priming of virus-specific T cells that can protect against IAV even in the absence of preexisting neutralizing antibodies. CD4 T cells represent a particularly attractive target as they help to promote responses by other innate and adaptive lymphocyte populations and can also directly mediate potent effector functions. Studies in murine models of IAV infection have been instrumental in moving this goal forward. Here, we will review these findings, focusing on distinct subsets of CD4 T cell effectors that have been shown to impact outcomes. This body of work suggests that a major challenge for next-generation vaccines will be to prime a CD4 T cell population with the same spectrum of functional diversity generated by IAV infection. This goal is encapsulated well by the motto 'ex pluribus unum': that an optimal CD4 T cell response comprises many individual specialized subsets responding together.
Collapse
Affiliation(s)
| | - K. Kai McKinstry
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA;
| |
Collapse
|
3
|
West EE, Merle NS, Kamiński MM, Palacios G, Kumar D, Wang L, Bibby JA, Overdahl K, Jarmusch AK, Freeley S, Lee DY, Thompson JW, Yu ZX, Taylor N, Sitbon M, Green DR, Bohrer A, Mayer-Barber KD, Afzali B, Kazemian M, Scholl-Buergi S, Karall D, Huemer M, Kemper C. Loss of CD4 + T cell-intrinsic arginase 1 accelerates Th1 response kinetics and reduces lung pathology during influenza infection. Immunity 2023; 56:2036-2053.e12. [PMID: 37572656 PMCID: PMC10576612 DOI: 10.1016/j.immuni.2023.07.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 06/01/2023] [Accepted: 07/19/2023] [Indexed: 08/14/2023]
Abstract
Arginase 1 (Arg1), the enzyme catalyzing the conversion of arginine to ornithine, is a hallmark of IL-10-producing immunoregulatory M2 macrophages. However, its expression in T cells is disputed. Here, we demonstrate that induction of Arg1 expression is a key feature of lung CD4+ T cells during mouse in vivo influenza infection. Conditional ablation of Arg1 in CD4+ T cells accelerated both virus-specific T helper 1 (Th1) effector responses and its resolution, resulting in efficient viral clearance and reduced lung pathology. Using unbiased transcriptomics and metabolomics, we found that Arg1-deficiency was distinct from Arg2-deficiency and caused altered glutamine metabolism. Rebalancing this perturbed glutamine flux normalized the cellular Th1 response. CD4+ T cells from rare ARG1-deficient patients or CRISPR-Cas9-mediated ARG1-deletion in healthy donor cells phenocopied the murine cellular phenotype. Collectively, CD4+ T cell-intrinsic Arg1 functions as an unexpected rheostat regulating the kinetics of the mammalian Th1 lifecycle with implications for Th1-associated tissue pathologies.
Collapse
Affiliation(s)
- Erin E West
- Complement and Inflammation Research Section (CIRS), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA.
| | - Nicolas S Merle
- Complement and Inflammation Research Section (CIRS), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Marcin M Kamiński
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gustavo Palacios
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Dhaneshwar Kumar
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Luopin Wang
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, USA
| | - Jack A Bibby
- Complement and Inflammation Research Section (CIRS), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Kirsten Overdahl
- Immunity, Inflammation, and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, NC, USA
| | - Alan K Jarmusch
- Immunity, Inflammation, and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, NC, USA
| | - Simon Freeley
- School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | | | - J Will Thompson
- Proteomics and Metabolomics Shared Resource, Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Zu-Xi Yu
- Pathology Core, NHLBI, NIH, Bethesda, MD, USA
| | - Naomi Taylor
- Pediatric Oncology Branch, Rare Tumor Initiative, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, USA; Institut de Génétique Moléculaire de Montpellier (IGMM), Université de Montpellier, CNRS, Montpellier, France
| | - Marc Sitbon
- Pediatric Oncology Branch, Rare Tumor Initiative, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, USA; Institut de Génétique Moléculaire de Montpellier (IGMM), Université de Montpellier, CNRS, Montpellier, France
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Andrea Bohrer
- Inflammation and Innate Immunity Unit, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, USA
| | - Katrin D Mayer-Barber
- Inflammation and Innate Immunity Unit, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, USA
| | - Behdad Afzali
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Majid Kazemian
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, USA
| | - Sabine Scholl-Buergi
- Clinic for Pediatrics I, Inherited Metabolic Disorders, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniela Karall
- Clinic for Pediatrics I, Inherited Metabolic Disorders, Medical University of Innsbruck, Innsbruck, Austria
| | - Martina Huemer
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland; Department of Pediatric Endocrinology and Diabetology, University Children's Hospital Basel, Basel, Switzerland; Department of Pediatrics, Landeskrankenhaus (LKH) Bregenz, Bregenz, Austria
| | - Claudia Kemper
- Complement and Inflammation Research Section (CIRS), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
4
|
Finney GE, Hargrave KE, Pingen M, Purnell T, Todd D, MacDonald F, Worrell JC, MacLeod MKL. Triphasic production of IFN γ by innate and adaptive lymphocytes following influenza A virus infection. DISCOVERY IMMUNOLOGY 2023; 2:kyad014. [PMID: 37842651 PMCID: PMC10568397 DOI: 10.1093/discim/kyad014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/02/2023] [Accepted: 08/09/2023] [Indexed: 10/17/2023]
Abstract
Interferon gamma (IFNγ) is a potent antiviral cytokine that can be produced by many innate and adaptive immune cells during infection. Currently, our understanding of which cells produce IFNγ and where they are located at different stages of an infection is limited. We have used reporter mice to investigate in vivo expression of Ifnγ mRNA in the lung and secondary lymphoid organs during and following influenza A virus (IAV) infection. We observed a triphasic production of Ifnγ expression. Unconventional T cells and innate lymphoid cells, particularly NK cells, were the dominant producers of early Ifnγ, while CD4 and CD8 T cells were the main producers by day 10 post-infection. Following viral clearance, some memory CD4 and CD8 T cells continued to express Ifnγ in the lungs and draining lymph node. Interestingly, Ifnγ production by lymph node natural killer (NK), NKT, and innate lymphoid type 1 cells also continued to be above naïve levels, suggesting memory-like phenotypes for these cells. Analysis of the localization of Ifnγ+ memory CD4 and CD8 T cells demonstrated that cytokine+ T cells were located near airways and in the lung parenchyma. Following a second IAV challenge, lung IAV-specific CD8 T cells rapidly increased their expression of Ifnγ while CD4 T cells in the draining lymph node increased their Ifnγ response. Together, these data suggest that Ifnγ production fluctuates based on cellular source and location, both of which could impact subsequent immune responses.
Collapse
Affiliation(s)
- George E Finney
- Centre for Immunobiology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Kerrie E Hargrave
- Centre for Immunobiology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Marieke Pingen
- Centre for Immunobiology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Thomas Purnell
- Centre for Immunobiology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - David Todd
- Centre for Immunobiology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Freya MacDonald
- Centre for Immunobiology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Julie C Worrell
- Centre for Immunobiology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Megan K L MacLeod
- Centre for Immunobiology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
| |
Collapse
|
5
|
Krishnan R, Stapledon CJM, Mostafavi H, Freitas JR, Liu X, Mahalingam S, Zaid A. Anti-inflammatory actions of Pentosan polysulfate sodium in a mouse model of influenza virus A/PR8/34-induced pulmonary inflammation. Front Immunol 2023; 14:1030879. [PMID: 36845136 PMCID: PMC9947849 DOI: 10.3389/fimmu.2023.1030879] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Introduction There is an unmet medical need for effective anti-inflammatory agents for the treatment of acute and post-acute lung inflammation caused by respiratory viruses. The semi-synthetic polysaccharide, Pentosan polysulfate sodium (PPS), an inhibitor of NF-kB activation, was investigated for its systemic and local anti-inflammatory effects in a mouse model of influenza virus A/PR8/1934 (PR8 strain) mediated infection. Methods Immunocompetent C57BL/6J mice were infected intranasally with a sublethal dose of PR8 and treated subcutaneously with 3 or 6 mg/kg PPS or vehicle. Disease was monitored and tissues were collected at the acute (8 days post-infection; dpi) or post-acute (21 dpi) phase of disease to assess the effect of PPS on PR8-induced pathology. Results In the acute phase of PR8 infection, PPS treatment was associated with a reduction in weight loss and improvement in oxygen saturation when compared to vehicle-treated mice. Associated with these clinical improvements, PPS treatment showed a significant retention in the numbers of protective SiglecF+ resident alveolar macrophages, despite uneventful changes in pulmonary leukocyte infiltrates assessed by flow cytometry. PPS treatment in PR8- infected mice showed significant reductions systemically but not locally of the inflammatory molecules, IL-6, IFN-g, TNF-a, IL-12p70 and CCL2. In the post-acute phase of infection, PPS demonstrated a reduction in the pulmonary fibrotic biomarkers, sICAM-1 and complement factor C5b9. Discussion The systemic and local anti-inflammatory actions of PPS may regulate acute and post-acute pulmonary inflammation and tissue remodeling mediated by PR8 infection, which warrants further investigation.
Collapse
Affiliation(s)
- Ravi Krishnan
- Research and Development, Paradigm Biopharmaceuticals Ltd., Melbourne, VIC, Australia
| | | | - Helen Mostafavi
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
- Global Virus Network (GVN) Center for Excellence in Arboviruses, Griffith University, Gold Coast, QLD, Australia
| | - Joseph R. Freitas
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
- Global Virus Network (GVN) Center for Excellence in Arboviruses, Griffith University, Gold Coast, QLD, Australia
| | - Xiang Liu
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
- Global Virus Network (GVN) Center for Excellence in Arboviruses, Griffith University, Gold Coast, QLD, Australia
| | - Suresh Mahalingam
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
- Global Virus Network (GVN) Center for Excellence in Arboviruses, Griffith University, Gold Coast, QLD, Australia
| | - Ali Zaid
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
- Global Virus Network (GVN) Center for Excellence in Arboviruses, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
6
|
Recombinant Protein Vaccines Formulated with Enantio-Specific Cationic Lipid R-DOTAP Induce Protective Cellular and Antibody-Mediated Immune Responses in Mice. Viruses 2023; 15:v15020432. [PMID: 36851646 PMCID: PMC9965888 DOI: 10.3390/v15020432] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/08/2023] Open
Abstract
Adjuvants are essential components of subunit vaccines added to enhance immune responses to antigens through immunomodulation. Very few adjuvants have been approved for human use by regulatory agencies due to safety concerns. Current subunit vaccine adjuvants approved for human use are very effective in promoting humoral immune responses but are less effective at promoting T-cell immunity. In this study, we evaluated a novel pure enantio-specific cationic lipid 1,2-dioleoyl-3-trimethylammonium-propane (R-DOTAP) as an immunomodulator for subunit vaccines capable of inducing both humoral- and cellular-mediated immunity. Using recombinant protein antigens derived from SARS-CoV2 spike or novel computationally optimized broadly reactive influenza antigen (COBRA) proteins, we demonstrated that R-DOTAP nanoparticles promoted strong cellular- and antibody-mediated immune responses in both monovalent and bivalent vaccines. R-DOTAP-based vaccines induced antigen-specific and polyfunctional CD8+ and CD4+ effector T cells and memory T cells, respectively. Antibody responses induced by R-DOTAP showed a balanced Th1/Th2 type immunity, neutralizing activity and protection of mice from challenge with live SARS-CoV2 or influenza viruses. R-DOTAP also facilitated significant dose sparing of the vaccine antigens. These studies demonstrate that R-DOTAP is an excellent immune stimulator for the production of next-generation subunit vaccines containing multiple recombinant proteins.
Collapse
|
7
|
Abstract
Barrier tissues are the primary site of infection for pathogens likely to cause future pandemics. Tissue-resident lymphocytes can rapidly detect pathogens upon infection of barrier tissues and are critical in preventing viral spread. However, most vaccines fail to induce tissue-resident lymphocytes and are instead reliant on circulating antibodies to mediate protective immunity. Circulating antibody titers wane over time following vaccination leaving individuals susceptible to breakthrough infections by variant viral strains that evade antibody neutralization. Memory B cells were recently found to establish tissue residence following infection of barrier tissues. Here, we summarize emerging evidence for the importance of tissue-resident memory B cells in the establishment of protective immunity against viral and bacterial challenge. We also discuss the role of tissue-resident memory B cells in regulating the progression of non-infectious diseases. Finally, we examine new approaches to develop vaccines capable of eliciting barrier immunity.
Collapse
Affiliation(s)
- Changfeng Chen
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Brian J Laidlaw
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States.
| |
Collapse
|
8
|
Paolini A, Borella R, Neroni A, Lo Tartaro D, Mattioli M, Fidanza L, Di Nella A, Santacroce E, Gozzi L, Busani S, Trenti T, Meschiari M, Guaraldi G, Girardis M, Mussini C, Gibellini L, De Biasi S, Cossarizza A. Patients Recovering from Severe COVID-19 Develop a Polyfunctional Antigen-Specific CD4+ T Cell Response. Int J Mol Sci 2022; 23:8004. [PMID: 35887351 PMCID: PMC9323836 DOI: 10.3390/ijms23148004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 12/10/2022] Open
Abstract
Specific T cells are crucial to control SARS-CoV-2 infection, avoid reinfection and confer protection after vaccination. We have studied patients with severe or moderate COVID-19 pneumonia, compared to patients who recovered from a severe or moderate infection that had occurred about 4 months before the analyses. In all these subjects, we assessed the polyfunctionality of virus-specific CD4+ and CD8+ T cells by quantifying cytokine production after in vitro stimulation with different SARS-CoV-2 peptide pools covering different proteins (M, N and S). In particular, we quantified the percentage of CD4+ and CD8+ T cells simultaneously producing interferon-γ, tumor necrosis factor, interleukin (IL)-2, IL-17, granzyme B, and expressing CD107a. Recovered patients who experienced a severe disease display high proportions of antigen-specific CD4+ T cells producing Th1 and Th17 cytokines and are characterized by polyfunctional SARS-CoV-2-specific CD4+ T cells. A similar profile was found in patients experiencing a moderate form of COVID-19 pneumonia. No main differences in polyfunctionality were observed among the CD8+ T cell compartments, even if the proportion of responding cells was higher during the infection. The identification of those functional cell subsets that might influence protection can thus help in better understanding the complexity of immune response to SARS-CoV-2.
Collapse
Affiliation(s)
- Annamaria Paolini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Rebecca Borella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Anita Neroni
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Marco Mattioli
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Lucia Fidanza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Alessia Di Nella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Elena Santacroce
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Licia Gozzi
- Infectious Diseases Clinics, AOU Policlinico di Modena, Via del Pozzo 71, 41124 Modena, Italy; (L.G.); (M.M.); (G.G.); (C.M.)
| | - Stefano Busani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (S.B.); (M.G.)
- Department of Anesthesia and Intensive Care, AOU Policlinico and University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy;
| | - Marianna Meschiari
- Infectious Diseases Clinics, AOU Policlinico di Modena, Via del Pozzo 71, 41124 Modena, Italy; (L.G.); (M.M.); (G.G.); (C.M.)
| | - Giovanni Guaraldi
- Infectious Diseases Clinics, AOU Policlinico di Modena, Via del Pozzo 71, 41124 Modena, Italy; (L.G.); (M.M.); (G.G.); (C.M.)
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (S.B.); (M.G.)
| | - Massimo Girardis
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (S.B.); (M.G.)
- Department of Anesthesia and Intensive Care, AOU Policlinico and University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy
| | - Cristina Mussini
- Infectious Diseases Clinics, AOU Policlinico di Modena, Via del Pozzo 71, 41124 Modena, Italy; (L.G.); (M.M.); (G.G.); (C.M.)
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (S.B.); (M.G.)
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
- National Institute for Cardiovascular Research, Via Irnerio 48, 40126 Bologna, Italy
| |
Collapse
|
9
|
CD4 expression in effector T cells depends on DNA demethylation over a developmentally established stimulus-responsive element. Nat Commun 2022; 13:1477. [PMID: 35304452 PMCID: PMC8933563 DOI: 10.1038/s41467-022-28914-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 02/16/2022] [Indexed: 12/17/2022] Open
Abstract
The epigenetic patterns that are established during early thymic development might determine mature T cell physiology and function, but the molecular basis and topography of the genetic elements involved are not fully known. Here we show, using the Cd4 locus as a paradigm for early developmental programming, that DNA demethylation during thymic development licenses a novel stimulus-responsive element that is critical for the maintenance of Cd4 gene expression in effector T cells. We document the importance of maintaining high CD4 expression during parasitic infection and show that by driving transcription, this stimulus-responsive element allows for the maintenance of histone H3K4me3 levels during T cell replication, which is critical for preventing de novo DNA methylation at the Cd4 promoter. A failure to undergo epigenetic programming during development leads to gene silencing during effector T cell replication. Our study thus provides evidence of early developmental events shaping the functional fitness of mature effector T cells.
Collapse
|
10
|
Reed SG, Ager A. Immune Responses to IAV Infection and the Roles of L-Selectin and ADAM17 in Lymphocyte Homing. Pathogens 2022; 11:pathogens11020150. [PMID: 35215094 PMCID: PMC8878872 DOI: 10.3390/pathogens11020150] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/14/2022] [Accepted: 01/21/2022] [Indexed: 02/04/2023] Open
Abstract
Influenza A virus (IAV) infection is a global public health burden causing up to 650,000 deaths per year. Yearly vaccination programmes and anti-viral drugs currently have limited benefits; therefore, research into IAV is fundamental. Leukocyte trafficking is a crucial process which orchestrates the immune response to infection to protect the host. It involves several homing molecules and receptors on both blood vessels and leukocytes. A key mediator of this process is the transmembrane glycoprotein L-selectin, which binds to vascular addressins on blood vessel endothelial cells. L-selectin classically mediates homing of naïve and central memory lymphocytes to lymph nodes via high endothelial venules (HEVs). Recent studies have found that L-selectin is essential for homing of activated CD8+ T cells to influenza-infected lungs and reduction in virus load. A disintegrin and metalloproteinase 17 (ADAM17) is the primary regulator of cell surface levels of L-selectin. Understanding the mechanisms that regulate these two proteins are central to comprehending recruitment of T cells to sites of IAV infection. This review summarises the immune response to IAV infection in humans and mice and discusses the roles of L-selectin and ADAM17 in T lymphocyte homing during IAV infection.
Collapse
Affiliation(s)
| | - Ann Ager
- Correspondence: (S.G.R.); (A.A.)
| |
Collapse
|
11
|
Reijnders TDY, Schuurman AR, van der Poll T. The Immune Response to Respiratory Viruses: From Start to Memory. Semin Respir Crit Care Med 2021; 42:759-770. [PMID: 34918319 DOI: 10.1055/s-0041-1736459] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Biomedical research has long strived to improve our understanding of the immune response to respiratory viral infections, an effort that has become all the more important as we live through the consequences of a pandemic. The disease course of these infections is shaped in large part by the actions of various cells of the innate and adaptive immune systems. While these cells are crucial in clearing viral pathogens and establishing long-term immunity, their effector mechanisms may also escalate into excessive, tissue-destructive inflammation detrimental to the host. In this review, we describe the breadth of the immune response to infection with respiratory viruses such as influenza and respiratory syncytial virus. Throughout, we focus on the host rather than the pathogen and try to describe shared patterns in the host response to different viruses. We start with the local cells of the airways, onto the recruitment and activation of innate and adaptive immune cells, followed by the establishment of local and systemic memory cells key in protection against reinfection. We end by exploring how respiratory viral infections can predispose to bacterial superinfection.
Collapse
Affiliation(s)
- Tom D Y Reijnders
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Alex R Schuurman
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands.,Division of Infectious Diseases, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
12
|
Elfaki Y, Yang J, Boehme J, Schultz K, Bruder D, Falk CS, Huehn J, Floess S. Tbx21 and Foxp3 Are Epigenetically Stabilized in T-Bet + Tregs That Transiently Accumulate in Influenza A Virus-Infected Lungs. Int J Mol Sci 2021; 22:ijms22147522. [PMID: 34299148 PMCID: PMC8307036 DOI: 10.3390/ijms22147522] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 01/19/2023] Open
Abstract
During influenza A virus (IAV) infections, CD4+ T cell responses within infected lungs mainly involve T helper 1 (Th1) and regulatory T cells (Tregs). Th1-mediated responses favor the co-expression of T-box transcription factor 21 (T-bet) in Foxp3+ Tregs, enabling the efficient Treg control of Th1 responses in infected tissues. So far, the exact accumulation kinetics of T cell subsets in the lungs and lung-draining lymph nodes (dLN) of IAV-infected mice is incompletely understood, and the epigenetic signature of Tregs accumulating in infected lungs has not been investigated. Here, we report that the total T cell and the two-step Treg accumulation in IAV-infected lungs is transient, whereas the change in the ratio of CD4+ to CD8+ T cells is more durable. Within lungs, the frequency of Tregs co-expressing T-bet is steadily, yet transiently, increasing with a peak at Day 7 post-infection. Interestingly, T-bet+ Tregs accumulating in IAV-infected lungs displayed a strongly demethylated Tbx21 locus, similarly as in T-bet+ conventional T cells, and a fully demethylated Treg-specific demethylated region (TSDR) within the Foxp3 locus. In summary, our data suggest that T-bet+ but not T-bet- Tregs are epigenetically stabilized during IAV-induced infection in the lung.
Collapse
Affiliation(s)
- Yassin Elfaki
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (Y.E.); (J.Y.)
| | - Juhao Yang
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (Y.E.); (J.Y.)
| | - Julia Boehme
- Immune Regulation Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (J.B.); (K.S.); (D.B.)
| | - Kristin Schultz
- Immune Regulation Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (J.B.); (K.S.); (D.B.)
| | - Dunja Bruder
- Immune Regulation Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (J.B.); (K.S.); (D.B.)
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Christine S. Falk
- Institute of Transplant Immunology, Hannover Medical School, 30625 Hannover, Germany;
- German Center for Infection Research DZIF, Thematical Translation Unit-Immunocompromized Host (TTU-IICH), Hannover-Braunschweig Site, 30625 Hannover, Germany
| | - Jochen Huehn
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (Y.E.); (J.Y.)
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany
- Correspondence: (J.H.); (S.F.)
| | - Stefan Floess
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (Y.E.); (J.Y.)
- Correspondence: (J.H.); (S.F.)
| |
Collapse
|
13
|
Thomann AS, Schneider T, Cyran L, Eckert IN, Kerstan A, Lutz MB. Conversion of Anergic T Cells Into Foxp3 - IL-10 + Regulatory T Cells by a Second Antigen Stimulus In Vivo. Front Immunol 2021; 12:704578. [PMID: 34249012 PMCID: PMC8267912 DOI: 10.3389/fimmu.2021.704578] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/15/2021] [Indexed: 11/16/2022] Open
Abstract
T cell anergy is a common mechanism of T cell tolerance. However, although anergic T cells are retained for longer time periods in their hosts, they remain functionally passive. Here, we describe the induction of anergic CD4+ T cells in vivo by intravenous application of high doses of antigen and their subsequent conversion into suppressive Foxp3- IL-10+ Tr1 cells but not Foxp3+ Tregs. We describe the kinetics of up-regulation of several memory-, anergy- and suppression-related markers such as CD44, CD73, FR4, CD25, CD28, PD-1, Egr-2, Foxp3 and CTLA-4 in this process. The conversion into suppressive Tr1 cells correlates with the transient intracellular CTLA-4 expression and required the restimulation of anergic cells in a short-term time window. Restimulation after longer time periods, when CTLA-4 is down-regulated again retains the anergic state but does not lead to the induction of suppressor function. Our data require further functional investigations but at this stage may suggest a role for anergic T cells as a circulating pool of passive cells that may be re-activated into Tr1 cells upon short-term restimulation with high and systemic doses of antigen. It is tentative to speculate that such a scenario may represent cases of allergen responses in non-allergic individuals.
Collapse
Affiliation(s)
- Anna Sophie Thomann
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Theresa Schneider
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Laura Cyran
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Ina Nathalie Eckert
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Andreas Kerstan
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Manfred B Lutz
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
14
|
BOCCALINI SARA, PARIANI ELENA, CALABRÒ GIOVANNAELISA, DE WAURE CHIARA, PANATTO DONATELLA, AMICIZIA DANIELA, LAI PIEROLUIGI, RIZZO CATERINA, AMODIO EMANUELE, VITALE FRANCESCO, CASUCCIO ALESSANDRA, DI PIETRO MARIALUISA, GALLI CRISTINA, BUBBA LAURA, PELLEGRINELLI LAURA, VILLANI LEONARDO, D’AMBROSIO FLORIANA, CAMINITI MARTA, LORENZINI ELISA, FIORETTI PAOLA, MICALE ROSANNATINDARA, FRUMENTO DAVIDE, CANTOVA ELISA, PARENTE FLAVIO, TRENTO GIACOMO, SOTTILE SARA, PUGLIESE ANDREA, BIAMONTE MASSIMILIANOALBERTO, GIORGETTI DUCCIO, MENICACCI MARCO, D’ANNA ANTONIO, AMMOSCATO CLAUDIA, LA GATTA EMANUELE, BECHINI ANGELA, BONANNI PAOLO. [Health Technology Assessment (HTA) of the introduction of influenza vaccination for Italian children with Fluenz Tetra ®]. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2021; 62:E1-E118. [PMID: 34909481 PMCID: PMC8639053 DOI: 10.15167/2421-4248/jpmh2021.62.2s1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- SARA BOCCALINI
- Dipartimento di Scienze della Salute, Università degli Studi di Firenze, Firenze, Italia
| | - ELENA PARIANI
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milano, Italia
- Centro Interuniversitario per la Ricerca sull'Influenza e le altre Infezioni Trasmissibili CIRI-IT, Italia
| | - GIOVANNA ELISA CALABRÒ
- Sezione di Igiene, Dipartimento Universitario di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Roma, Italia
- VIHTALI (Value In Health Technology and Academy for Leadership & Innovation), spin off dell’Università Cattolica del Sacro Cuore, Roma, Italia
| | - CHIARA DE WAURE
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, Perugia, Italia
| | - DONATELLA PANATTO
- Centro Interuniversitario per la Ricerca sull'Influenza e le altre Infezioni Trasmissibili CIRI-IT, Italia
- Dipartimento di Scienze della Salute, Università degli Studi di Genova, Genova, Italia
| | - DANIELA AMICIZIA
- Centro Interuniversitario per la Ricerca sull'Influenza e le altre Infezioni Trasmissibili CIRI-IT, Italia
- Dipartimento di Scienze della Salute, Università degli Studi di Genova, Genova, Italia
| | - PIERO LUIGI LAI
- Centro Interuniversitario per la Ricerca sull'Influenza e le altre Infezioni Trasmissibili CIRI-IT, Italia
- Dipartimento di Scienze della Salute, Università degli Studi di Genova, Genova, Italia
| | - CATERINA RIZZO
- Area Funzionale Percorsi Clinici ed Epidemiologia, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italia
| | - EMANUELE AMODIO
- Dipartimento Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D'Alessandro”, Università degli Studi di Palermo, Palermo, Italia
| | - FRANCESCO VITALE
- Dipartimento Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D'Alessandro”, Università degli Studi di Palermo, Palermo, Italia
| | - ALESSANDRA CASUCCIO
- Dipartimento Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D'Alessandro”, Università degli Studi di Palermo, Palermo, Italia
| | - MARIA LUISA DI PIETRO
- Sezione di Igiene, Dipartimento Universitario di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - CRISTINA GALLI
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milano, Italia
| | - LAURA BUBBA
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milano, Italia
| | - LAURA PELLEGRINELLI
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milano, Italia
| | - LEONARDO VILLANI
- Sezione di Igiene, Dipartimento Universitario di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - FLORIANA D’AMBROSIO
- Sezione di Igiene, Dipartimento Universitario di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - MARTA CAMINITI
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, Perugia, Italia
| | - ELISA LORENZINI
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, Perugia, Italia
| | - PAOLA FIORETTI
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, Perugia, Italia
| | | | - DAVIDE FRUMENTO
- Dipartimento di Scienze della Salute, Università degli Studi di Genova, Genova, Italia
| | - ELISA CANTOVA
- Dipartimento di Scienze della Salute, Università degli Studi di Genova, Genova, Italia
| | - FLAVIO PARENTE
- Dipartimento di Scienze della Salute, Università degli Studi di Genova, Genova, Italia
| | - GIACOMO TRENTO
- Dipartimento di Scienze della Salute, Università degli Studi di Genova, Genova, Italia
| | - SARA SOTTILE
- Università degli Studi di Trento, Trento, Italia
| | | | | | - DUCCIO GIORGETTI
- Dipartimento di Scienze della Salute, Università degli Studi di Firenze, Firenze, Italia
| | - MARCO MENICACCI
- Dipartimento di Scienze della Salute, Università degli Studi di Firenze, Firenze, Italia
| | - ANTONIO D’ANNA
- Dipartimento Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D'Alessandro”, Università degli Studi di Palermo, Palermo, Italia
| | - CLAUDIA AMMOSCATO
- Dipartimento Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D'Alessandro”, Università degli Studi di Palermo, Palermo, Italia
| | - EMANUELE LA GATTA
- Sezione di Igiene, Dipartimento Universitario di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - ANGELA BECHINI
- Dipartimento di Scienze della Salute, Università degli Studi di Firenze, Firenze, Italia
| | - PAOLO BONANNI
- Dipartimento di Scienze della Salute, Università degli Studi di Firenze, Firenze, Italia
| |
Collapse
|
15
|
Son YM, Cheon IS, Wu Y, Li C, Wang Z, Gao X, Chen Y, Takahashi Y, Fu YX, Dent AL, Kaplan MH, Taylor JJ, Cui W, Sun J. Tissue-resident CD4 + T helper cells assist the development of protective respiratory B and CD8 + T cell memory responses. Sci Immunol 2021; 6:6/55/eabb6852. [PMID: 33419791 DOI: 10.1126/sciimmunol.abb6852] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 11/11/2020] [Indexed: 11/02/2022]
Abstract
Much remains unknown about the roles of CD4+ T helper cells in shaping localized memory B cell and CD8+ T cell immunity in the mucosal tissues. Here, we report that lung T helper cells provide local assistance for the optimal development of tissue-resident memory B and CD8+ T cells after the resolution of primary influenza virus infection. We have identified a population of T cells in the lung that exhibit characteristics of both follicular T helper and TRM cells, and we have termed these cells as resident helper T (TRH) cells. Optimal TRH cell formation was dependent on transcription factors involved in T follicular helper and resident memory T cell development including BCL6 and Bhlhe40. We show that TRH cells deliver local help to CD8+ T cells through IL-21-dependent mechanisms. Our data have uncovered the presence of a tissue-resident helper T cell population in the lung that plays a critical role in promoting the development of protective B cell and CD8+ T cell responses.
Collapse
Affiliation(s)
- Young Min Son
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA.,Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - In Su Cheon
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA.,Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Yue Wu
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA.,Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Chaofan Li
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA.,Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Zheng Wang
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA.,Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Xiaochen Gao
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA.,Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Yao Chen
- Versiti Blood Research Institute, Milwaukee, WI 53213, USA
| | - Yoshimasa Takahashi
- Department of Immunology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Yang-Xin Fu
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Alexander L Dent
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Mark H Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Justin J Taylor
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Weiguo Cui
- Versiti Blood Research Institute, Milwaukee, WI 53213, USA.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jie Sun
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA. .,Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
16
|
Zaunders J, Phetsouphanh C. Long-term and short-term immunity to SARS-CoV-2: why it matters. MICROBIOLOGY AUSTRALIA 2021. [DOI: 10.1071/ma21010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The adaptive immune system, regulated by CD4 T cells, is essential for control of many viral infections. Endemic coronavirus infections generally occur as short-term upper respiratory tract infections which in many cases appear to be cleared before adaptive immunity is fully involved, since adaptive immunity takes approximately 1.5–2 weeks to ramp up the response to a primary infection, or approximately 1 week for a recurrent infection. However, the adaptive immune response to SARS-CoV-2 infection will be critical to full recovery with minimal long-lasting effects, and to either prevention of recurrence of infection or at least reduced severity of symptoms. The detailed kinetics of this infection versus the dynamics of the immune response, including in vaccinated individuals, will largely determine these outcomes.
Collapse
|
17
|
Limbic Encephalitis Brain Damage Induced by Cocal Virus in Adult Mice Is Reduced by Environmental Enrichment: Neuropathological and Behavioral Studies. Viruses 2020; 13:v13010048. [PMID: 33396704 PMCID: PMC7824630 DOI: 10.3390/v13010048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 12/14/2020] [Indexed: 12/26/2022] Open
Abstract
We previously demonstrated, using the Piry virus model, that environmental enrichment promotes higher T-cell infiltration, fewer microglial changes, and faster central nervous system (CNS) virus clearance in adult mice. However, little is known about disease progression, behavioral changes, CNS cytokine concentration, and neuropathology in limbic encephalitis in experimental models. Using Cocal virus, we infected C57Bl6 adult mice and studied the neuroanatomical distribution of viral antigens in correlation with the microglial morphological response, measured the CNS cytokine concentration, and assessed behavioral changes. C57Bl6 adult mice were maintained in an impoverished environment (IE) or enriched environment (EE) for four months and then subjected to the open field test. Afterwards, an equal volume of normal or virus-infected brain homogenate was nasally instilled. The brains were processed to detect viral antigens and microglial morphological changes using selective immunolabeling. We demonstrated earlier significant weight loss and higher mortality in IE mice. Additionally, behavioral analysis revealed a significant influence of the environment on locomotor and exploratory activity that was associated with less neuroinvasion and a reduced microglial response. Thus, environmental enrichment was associated with a more effective immune response in a mouse model of limbic encephalitis, allowing faster viral clearance/decreased viral dissemination, reduced disease progression, and less CNS damage.
Collapse
|
18
|
Lopez CE, Legge KL. Influenza A Virus Vaccination: Immunity, Protection, and Recent Advances Toward A Universal Vaccine. Vaccines (Basel) 2020; 8:E434. [PMID: 32756443 PMCID: PMC7565301 DOI: 10.3390/vaccines8030434] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/28/2020] [Accepted: 07/31/2020] [Indexed: 12/19/2022] Open
Abstract
Influenza virus infections represent a serious public health threat and account for significant morbidity and mortality worldwide due to seasonal epidemics and periodic pandemics. Despite being an important countermeasure to combat influenza virus and being highly efficacious when matched to circulating influenza viruses, current preventative strategies of vaccination against influenza virus often provide incomplete protection due the continuous antigenic drift/shift of circulating strains of influenza virus. Prevention and control of influenza virus infection with vaccines is dependent on the host immune response induced by vaccination and the various vaccine platforms induce different components of the local and systemic immune response. This review focuses on the immune basis of current (inactivated influenza vaccines (IIV) and live attenuated influenza vaccines (LAIV)) as well as novel vaccine platforms against influenza virus. Particular emphasis will be placed on how each platform induces cross-protection against heterologous influenza viruses, as well as how this immunity compares to and contrasts from the "gold standard" of immunity generated by natural influenza virus infection.
Collapse
Affiliation(s)
- Christopher E. Lopez
- Department of Microbiology and Immunology University of Iowa, Iowa City, IA 52242, USA;
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Kevin L. Legge
- Department of Microbiology and Immunology University of Iowa, Iowa City, IA 52242, USA;
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
19
|
Host-Virus Interaction: How Host Cells Defend against Influenza A Virus Infection. Viruses 2020; 12:v12040376. [PMID: 32235330 PMCID: PMC7232439 DOI: 10.3390/v12040376] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/19/2020] [Accepted: 03/25/2020] [Indexed: 02/07/2023] Open
Abstract
Influenza A viruses (IAVs) are highly contagious pathogens infecting human and numerous animals. The viruses cause millions of infection cases and thousands of deaths every year, thus making IAVs a continual threat to global health. Upon IAV infection, host innate immune system is triggered and activated to restrict virus replication and clear pathogens. Subsequently, host adaptive immunity is involved in specific virus clearance. On the other hand, to achieve a successful infection, IAVs also apply multiple strategies to avoid be detected and eliminated by the host immunity. In the current review, we present a general description on recent work regarding different host cells and molecules facilitating antiviral defenses against IAV infection and how IAVs antagonize host immune responses.
Collapse
|