1
|
Liu L, Yi G, Li X, Chen C, Chen K, He H, Li J, Cai F, Peng Y, Yang Z, Zhang X. IL-17A's role in exacerbating radiation-induced lung injury: Autophagy impairment via the PP2A-mTOR pathway. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1872:119864. [PMID: 39437853 DOI: 10.1016/j.bbamcr.2024.119864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 09/17/2024] [Accepted: 10/12/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVE Radiation-induced lung injury (RILI) is a serious complication of radiotherapy, and the role of IL-17A in this process is not well understood. While IL-17A has been shown to modulate autophagy, conflicting reports exist regarding its activation or inhibition of autophagy. This study investigates the role of IL-17A in RILI and its effects on autophagy via the PP2A-mTOR pathway, with a focus on the PP2A B56α subunit. METHODS C57BL/6J mice and human lung epithelial cells (BEAS-2B) were exposed to radiation with or without recombinant IL-17A. Autophagy markers were analyzed using Western blotting, immunofluorescence, and autophagy flux assays. PP2A activity, specifically the B56α subunit, was measured. A PP2A agonist (DT-061) was used to verify its role in reversing IL-17A-mediated autophagy inhibition. RESULTS IL-17A inhibited autophagy in lung epithelial cells exposed to radiation by suppressing PP2A activity, particularly through downregulation of the B56α subunit, leading to mTOR activation and reduced autophagosome formation. Treatment with DT-061 restored autophagic activity and improved cell viability. These findings align with reports suggesting that IL-17A inhibits autophagy in certain contexts, while other studies have shown opposing effects. CONCLUSION IL-17A inhibits autophagy in RILI through the PP2A B56α-mTOR pathway, exacerbating lung damage. Further research is needed to clarify the role of IL-17A in different cell types and conditions. Targeting the IL-17A-PP2A B56α-mTOR axis may offer new therapeutic strategies for RILI management.
Collapse
Affiliation(s)
- Liangzhong Liu
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China; Department of Oncology, Chongqing University Three Gorges Hospital, Chongqing University, Chongqing 404100, China
| | - GuangMing Yi
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xiaohong Li
- Nursing Department, Chongqing University Three Gorges Hospital, Chongqing University, Chongqing 404100, China
| | - Cai Chen
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Kehong Chen
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Hengqiu He
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jinjin Li
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Fanghao Cai
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yuan Peng
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Zhenzhou Yang
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Xiaoyue Zhang
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| |
Collapse
|
2
|
Auld SC, Sheshadri A, Alexander-Brett J, Aschner Y, Barczak AK, Basil MC, Cohen KA, Dela Cruz C, McGroder C, Restrepo MI, Ridge KM, Schnapp LM, Traber K, Wunderink RG, Zhang D, Ziady A, Attia EF, Carter J, Chalmers JD, Crothers K, Feldman C, Jones BE, Kaminski N, Keane J, Lewinsohn D, Metersky M, Mizgerd JP, Morris A, Ramirez J, Samarasinghe AE, Staitieh BS, Stek C, Sun J, Evans SE. Postinfectious Pulmonary Complications: Establishing Research Priorities to Advance the Field: An Official American Thoracic Society Workshop Report. Ann Am Thorac Soc 2024; 21:1219-1237. [PMID: 39051991 DOI: 10.1513/annalsats.202406-651st] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Continued improvements in the treatment of pulmonary infections have paradoxically resulted in a growing challenge of individuals with postinfectious pulmonary complications (PIPCs). PIPCs have been long recognized after tuberculosis, but recent experiences such as the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic have underscored the importance of PIPCs following other lower respiratory tract infections. Independent of the causative pathogen, most available studies of pulmonary infections focus on short-term outcomes rather than long-term morbidity among survivors. In this document, we establish a conceptual scope for PIPCs with discussion of globally significant pulmonary pathogens and an examination of how these pathogens can damage different components of the lung, resulting in a spectrum of PIPCs. We also review potential mechanisms for the transition from acute infection to PIPC, including the interplay between pathogen-mediated injury and aberrant host responses, which together result in PIPCs. Finally, we identify cross-cutting research priorities for the field to facilitate future studies to establish the incidence of PIPCs, define common mechanisms, identify therapeutic strategies, and ultimately reduce the burden of morbidity in survivors of pulmonary infections.
Collapse
|
3
|
Korkmaz FT, Quinton LJ. Extra-pulmonary control of respiratory defense. Cell Immunol 2024; 401-402:104841. [PMID: 38878619 DOI: 10.1016/j.cellimm.2024.104841] [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: 03/03/2024] [Accepted: 06/06/2024] [Indexed: 07/13/2024]
Abstract
Pneumonia persists as a public health crisis, representing the leading cause of death due to infection. Whether respiratory tract infections progress to pneumonia and its sequelae such as acute respiratory distress syndrome and sepsis depends on numerous underlying conditions related to both the causative agent and host. Regarding the former, pneumonia burden remains staggeringly high, despite the effectiveness of pathogen-targeting strategies such as vaccines and antibiotics. This demands a greater understanding of host features that collaborate to promote immune resistance and tissue resilience in the infected lung. Such features inside the pulmonary compartment have drawn much attention, where major advances have been made related to resident and recruited immune activity. By comparison, extra-pulmonary processes guiding pneumonia susceptibility are relatively elusive, constituting the focus of this review. Here we will highlight examples of when, how, and why tissues outside of the lungs dispatch signals that modulate local immunity in the airspaces. Topics include the liver, gut, bone marrow, brain and more, all of which contribute in direct and indirect ways to pneumonia outcome. When tuned appropriately, it has become clear that these responses can serve protective roles, and this will be considered distinctly from what would otherwise be aberrant responses characteristic of pneumonia-induced organ injury and sepsis. Further advances in this area may reveal novel targetable areas for clinical intervention that are not confined to the intra-pulmonary space.
Collapse
Affiliation(s)
- Filiz T Korkmaz
- Department of Medicine, Division of Immunology and Infectious Disease, UMass Chan Medical School, Worcester, MA 01602, United States.
| | - Lee J Quinton
- Department of Medicine, Division of Immunology and Infectious Disease, UMass Chan Medical School, Worcester, MA 01602, United States
| |
Collapse
|
4
|
Etesami NS, Barker KA, Shenoy AT, De Ana CL, Arafa EI, Grifno GN, Matschulat AM, Vannini ME, Pihl RMF, Breen MP, Soucy AM, Goltry WN, Ha CT, Betsuyaku H, Browning JL, Varelas X, Traber KE, Jones MR, Quinton LJ, Maglione PJ, Nia HT, Belkina AC, Mizgerd JP. B cells in the pneumococcus-infected lung are heterogeneous and require CD4 + T cell help including CD40L to become resident memory B cells. Front Immunol 2024; 15:1382638. [PMID: 38715601 PMCID: PMC11074383 DOI: 10.3389/fimmu.2024.1382638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/01/2024] [Indexed: 05/12/2024] Open
Abstract
Recovery from respiratory pneumococcal infections generates lung-localized protection against heterotypic bacteria, mediated by resident memory lymphocytes. Optimal protection in mice requires re-exposure to pneumococcus within days of initial infection. Serial surface marker phenotyping of B cell populations in a model of pneumococcal heterotypic immunity revealed that bacterial re-exposure stimulates the immediate accumulation of dynamic and heterogeneous populations of B cells in the lung, and is essential for the establishment of lung resident memory B (BRM) cells. The B cells in the early wave were activated, proliferating locally, and associated with both CD4+ T cells and CXCL13. Antagonist- and antibody-mediated interventions were implemented during this early timeframe to demonstrate that lymphocyte recirculation, CD4+ cells, and CD40 ligand (CD40L) signaling were all needed for lung BRM cell establishment, whereas CXCL13 signaling was not. While most prominent as aggregates in the loose connective tissue of bronchovascular bundles, morphometry and live lung imaging analyses showed that lung BRM cells were equally numerous as single cells dispersed throughout the alveolar septae. We propose that CD40L signaling from antigen-stimulated CD4+ T cells in the infected lung is critical to establishment of local BRM cells, which subsequently protect the airways and parenchyma against future potential infections.
Collapse
Affiliation(s)
- Neelou S. Etesami
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Kimberly A. Barker
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Anukul T. Shenoy
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Carolina Lyon De Ana
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Emad I. Arafa
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Gabrielle N. Grifno
- Department of Biomedical Engineering, Boston University College of Engineering, Boston, MA, United States
| | - Adeline M. Matschulat
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Michael E. Vannini
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Riley M. F. Pihl
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Michael P. Breen
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Alicia M. Soucy
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Wesley N. Goltry
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Catherine T. Ha
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Hanae Betsuyaku
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Jeffrey L. Browning
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Xaralabos Varelas
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Katrina E. Traber
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Matthew R. Jones
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Lee J. Quinton
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Division of Infectious Diseases and Immunology, University of Massachusetts Chan Medical School, Worcester, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Paul J. Maglione
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Hadi T. Nia
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biomedical Engineering, Boston University College of Engineering, Boston, MA, United States
| | - Anna C. Belkina
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Flow Cytometry Core Facility, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Joseph P. Mizgerd
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| |
Collapse
|
5
|
Murakami M. Tissue-resident memory T cells: decoding intra-organ diversity with a gut perspective. Inflamm Regen 2024; 44:19. [PMID: 38632596 PMCID: PMC11022361 DOI: 10.1186/s41232-024-00333-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/05/2024] [Indexed: 04/19/2024] Open
Abstract
Tissue-resident memory T cells (TRM) serve as the frontline of host defense, playing a critical role in protection against invading pathogens. This emphasizes their role in providing rapid on-site immune responses across various organs. The physiological significance of TRM is not just confined to infection control; accumulating evidence has revealed that TRM also determine the pathology of diseases such as autoimmune disorders, inflammatory bowel disease, and cancer. Intensive studies on the origin, mechanisms of formation and maintenance, and physiological significance of TRM have elucidated the transcriptional and functional diversity of these cells, which are often affected by local cues associated with their presence. These were further confirmed by the recent remarkable advancements of next-generation sequencing and single-cell technologies, which allow the transcriptional and phenotypic characterization of each TRM subset induced in different microenvironments. This review first overviews the current knowledge of the cell fate, molecular features, transcriptional and metabolic regulation, and biological importance of TRM in health and disease. Finally, this article presents a variety of recent studies on disease-associated TRM, particularly focusing and elaborating on the TRM in the gut, which constitute the largest and most intricate immune network in the body, and their pathological relevance to gut inflammation in humans.
Collapse
Affiliation(s)
- Mari Murakami
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka, 565-0871, Japan.
- Immunology Frontier Research Center, Osaka University, Osaka, 565-0871, Japan.
| |
Collapse
|
6
|
Liu X, Van Maele L, Matarazzo L, Soulard D, Alves Duarte da Silva V, de Bakker V, Dénéréaz J, Bock FP, Taschner M, Ou J, Gruber S, Nizet V, Sirard JC, Veening JW. A conserved antigen induces respiratory Th17-mediated broad serotype protection against pneumococcal superinfection. Cell Host Microbe 2024; 32:304-314.e8. [PMID: 38417443 DOI: 10.1016/j.chom.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 12/06/2023] [Accepted: 02/05/2024] [Indexed: 03/01/2024]
Abstract
Several vaccines targeting bacterial pathogens show reduced efficacy upon concurrent viral infection, indicating that a new vaccinology approach is required. To identify antigens for the human pathogen Streptococcus pneumoniae that are effective following influenza infection, we performed CRISPRi-seq in a murine model of superinfection and identified the conserved lafB gene as crucial for virulence. We show that LafB is a membrane-associated, intracellular protein that catalyzes the formation of galactosyl-glucosyl-diacylglycerol, a glycolipid important for cell wall homeostasis. Respiratory vaccination with recombinant LafB, in contrast to subcutaneous vaccination, was highly protective against S. pneumoniae serotypes 2, 15A, and 24F in a murine model. In contrast to standard capsule-based vaccines, protection did not require LafB-specific antibodies but was dependent on airway CD4+ T helper 17 cells. Healthy human individuals can elicit LafB-specific immune responses, indicating LafB antigenicity in humans. Collectively, these findings present a universal pneumococcal vaccine antigen that remains effective following influenza infection.
Collapse
Affiliation(s)
- Xue Liu
- Department of Pathogen Biology, Base for International Science and Technology Cooperation, Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Medical School, Shenzhen 518060, China; Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, CH-1015 Lausanne, Switzerland
| | - Laurye Van Maele
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Laura Matarazzo
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Daphnée Soulard
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Vinicius Alves Duarte da Silva
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Vincent de Bakker
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, CH-1015 Lausanne, Switzerland
| | - Julien Dénéréaz
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, CH-1015 Lausanne, Switzerland
| | - Florian P Bock
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, CH-1015 Lausanne, Switzerland
| | - Michael Taschner
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, CH-1015 Lausanne, Switzerland
| | - Jinzhao Ou
- Department of Pathogen Biology, Base for International Science and Technology Cooperation, Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Medical School, Shenzhen 518060, China
| | - Stephan Gruber
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, CH-1015 Lausanne, Switzerland
| | - Victor Nizet
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Jean-Claude Sirard
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France.
| | - Jan-Willem Veening
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, CH-1015 Lausanne, Switzerland; Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
7
|
Trentini MM, Rodriguez D, Kanno AI, Goulart C, Darrieux M, de Cerqueira Leite LC. Robust Immune Response and Protection against Lethal Pneumococcal Challenge with a Recombinant BCG-PspA-PdT Prime/Boost Scheme Administered to Neonatal Mice. Vaccines (Basel) 2024; 12:122. [PMID: 38400107 PMCID: PMC10893189 DOI: 10.3390/vaccines12020122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Pneumococcal diseases are an important public health problem, with high mortality rates in young children. Although conjugated pneumococcal vaccines offer high protection against invasive pneumococcal diseases, this is restricted to vaccine serotypes, leading to serotype replacement. Furthermore, the current vaccines do not protect neonates. Therefore, several protein-based pneumococcal vaccines have been studied over the last few decades. Our group established a recombinant BCG expressing rPspA-PdT as a prime/rPspA-PdT boost strategy, which protected adult mice against lethal intranasal pneumococcal challenge. Here, we immunized groups of neonate C57/Bl6 mice (6-10) (at 5 days) with rBCG PspA-PdT and a boost with rPspA-PdT (at 12 days). Controls were saline or each antigen alone. The prime/boost strategy promoted an IgG1 to IgG2c isotype shift compared to protein alone. Furthermore, there was an increase in specific memory cells (T and B lymphocytes) and higher cytokine production (IFN-γ, IL-17, TNF-α, IL-10, and IL-6). Immunization with rBCG PspA-PdT/rPspA-PdT showed 100% protection against pulmonary challenge with the WU2 pneumococcal strain; two doses of rPspA-PdT showed non-significant protection in the neonates. These results demonstrate that a prime/boost strategy using rBCG PspA-PdT/rPspA-PdT is effective in protecting neonates against lethal pneumococcal infection via the induction of strong antibody and cytokine responses.
Collapse
Affiliation(s)
| | - Dunia Rodriguez
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Alex Issamu Kanno
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Cibelly Goulart
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Michelle Darrieux
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista 12916-900, Brazil;
| | | |
Collapse
|
8
|
Pei X, Liu L, Wang J, Guo C, Li Q, Li J, Ren Q, Ma R, Zheng Y, Zhang Y, Liu L, Zheng D, Wang P, Jiang P, Feng X, Jiang E, Wang Y, Feng S. Exosomal secreted SCIMP regulates communication between macrophages and neutrophils in pneumonia. Nat Commun 2024; 15:691. [PMID: 38263143 PMCID: PMC10805922 DOI: 10.1038/s41467-024-44714-4] [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: 04/11/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024] Open
Abstract
In pneumonia, the deficient or delayed pathogen clearance can lead to pathogen proliferation and subsequent overactive immune responses, inducing acute lung injury (ALI). While screening human genome coding genes using our peripheral blood cell chemotactic platform, we unexpectedly find SLP adaptor and CSK interacting membrane protein (SCIMP), a protein with neutrophil chemotactic activity secreted during ALI. However, the specific role of SCIMP in ALI remains unclear. In this study, we investigate the secretion of SCIMP in exosomes (SCIMPexo) by macrophages after bacterial stimulation, both in vitro and in vivo. We observe a significant increase in the levels of SCIMPexo in bronchoalveolar lavage fluid and serum of pneumonia patients. We also find that bronchial perfusion with SCIMPexo or SCIMP N-terminal peptides increases the survival rate of the ALI model. This occurs due to the chemoattraction and activation of peripheral neutrophils dependent on formyl peptide receptor 1/2 (FPR1/2). Conversely, exosome suppressors and FPR1/2 antagonists decrease the survival rate in the lethal ALI model. Scimp-deficient and Fpr1/2-deficient mice also have lower survival rates and shorter survival times than wild-type mice. However, bronchial perfusion of SCIMP rescues Scimp-deficient mice but not Fpr1/2-deficient mice. Collectively, our findings suggest that the macrophage-SCIMP-FPRs-neutrophil axis plays a vital role in the innate immune process underlying ALI.
Collapse
Affiliation(s)
- Xiaolei Pei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, P. R. China.
- Tianjin Institutes of Health Science, Tianjin, 301600, P. R. China.
| | - Li Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, P. R. China
- Tianjin Institutes of Health Science, Tianjin, 301600, P. R. China
| | - Jieru Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, P. R. China
- Tianjin Institutes of Health Science, Tianjin, 301600, P. R. China
| | - Changyuan Guo
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, P. R. China
| | - Qingqing Li
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, P. R. China
| | - Jia Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, P. R. China
- Tianjin Institutes of Health Science, Tianjin, 301600, P. R. China
| | - Qian Ren
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, P. R. China
- Tianjin Institutes of Health Science, Tianjin, 301600, P. R. China
| | - Runzhi Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, P. R. China
- Tianjin Institutes of Health Science, Tianjin, 301600, P. R. China
| | - Yi Zheng
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, P. R. China
| | - Yan Zhang
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, P. R. China
| | - Li Liu
- Tianjin First Central Hospital, Tianjin Medical University, Tianjin, 300192, P. R. China
| | - Danfeng Zheng
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, P. R. China
| | - Pingzhang Wang
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, P. R. China
| | - Ping Jiang
- Tianjin First Central Hospital, Tianjin Medical University, Tianjin, 300192, P. R. China
| | - Xiaoming Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, P. R. China
- Tianjin Institutes of Health Science, Tianjin, 301600, P. R. China
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, P. R. China
- Tianjin Institutes of Health Science, Tianjin, 301600, P. R. China
| | - Ying Wang
- Department of Immunology, School of Basic Medical Sciences and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, P. R. China.
| | - Sizhou Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, P. R. China.
- Tianjin Institutes of Health Science, Tianjin, 301600, P. R. China.
| |
Collapse
|
9
|
Zhang X, Zhang J, Chen S, He Q, Bai Y, Liu J, Wang Z, Liang Z, Chen L, Mao Q, Xu M. Progress and challenges in the clinical evaluation of immune responses to respiratory mucosal vaccines. Expert Rev Vaccines 2024; 23:362-370. [PMID: 38444382 DOI: 10.1080/14760584.2024.2326094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 02/28/2024] [Indexed: 03/07/2024]
Abstract
INTRODUCTION Following the coronavirus disease pandemic, respiratory mucosal vaccines that elicit both mucosal and systemic immune responses have garnered increasing attention. However, human physiological characteristics pose significant challenges in the evaluation of mucosal immunity, which directly impedes the development and application of respiratory mucosal vaccines. AREAS COVERED This study summarizes the characteristics of immune responses in the respiratory mucosa and reviews the current status and challenges in evaluating immune response to respiratory mucosal vaccines. EXPERT OPINION Secretory Immunoglobulin A (S-IgA) is a major effector molecule at mucosal sites and a commonly used indicator for evaluating respiratory mucosal vaccines. However, the unique physiological structure of the respiratory tract pose significant challenges for the clinical collection and detection of S-IgA. Therefore, it is imperative to develop a sampling method with high collection efficiency and acceptance, a sensitive detection method, reference materials for mucosal antibodies, and to establish a threshold for S-IgA that correlates with clinical protection. Sample collection is even more challenging when evaluating mucosal cell immunity. Therefore, a mucosal cell sampling method with high operability and high tolerance should be established. Targets of the circulatory system capable of reflecting mucosal cellular immunity should also be explored.
Collapse
Affiliation(s)
- Xuanxuan Zhang
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Jialu Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Si Chen
- Drug and Vaccine Research Center, Guangzhou National Laboratory, Guangzhou, China
| | - Qian He
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Yu Bai
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Jianyang Liu
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Zhongfang Wang
- Drug and Vaccine Research Center, Guangzhou National Laboratory, Guangzhou, China
| | - Zhenglun Liang
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Ling Chen
- Drug and Vaccine Research Center, Guangzhou National Laboratory, Guangzhou, China
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Qunying Mao
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Miao Xu
- State Key Laboratory of Drug Regulatory Science, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
10
|
Lyon De Ana C, Shenoy AT, Barker KA, Arafa EI, Etesami NS, Korkmaz FT, Soucy AM, Breen MP, Martin IMC, Tilton BR, Devarajan P, Crossland NA, Pihl RMF, Goltry WN, Belkina AC, Jones MR, Quinton LJ, Mizgerd JP. GL7 ligand expression defines a novel subset of CD4 + T RM cells in lungs recovered from pneumococcus. Mucosal Immunol 2023; 16:699-710. [PMID: 37604254 PMCID: PMC10591822 DOI: 10.1016/j.mucimm.2023.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 07/26/2023] [Indexed: 08/23/2023]
Abstract
Streptococcus pneumoniae is the most common etiology of bacterial pneumonia, one of the leading causes of death in children and the elderly worldwide. During non-lethal infections with S. pneumoniae, lymphocytes accumulate in the lungs and protect against reinfection with serotype-mismatched strains. Cluster of differentiation CD4+ resident memory T (TRM) cells are known to be crucial for this protection, but the diversity of lung CD4+ TRM cells has yet to be fully delineated. We aimed to identify unique subsets and their contributions to lung immunity. After recovery from pneumococcal infections, we identified a distinct subset of CD4+ T cells defined by the phenotype CD11ahiCD69+GL7+ in mouse lungs. Phenotypic analyses for markers of lymphocyte memory and residence demonstrated that GL7+ T cells are a subset of CD4+ TRM cells. Functional studies revealed that unlike GL7- TRM subsets that were mostly (RAR-related Orphan Receptor gamma T) RORγT+, GL7+ TRM cells exhibited higher levels of (T-box expressed in T cells) T-bet and Gata-3, corresponding with increased synthesis of interferon-γ, interleukin-13, and interleukin-5, inherent to both T helper 1 (TH1) and TH2 functions. Thus, we propose that these cells provide novel contributions during pneumococcal pneumonia, serving as important determinants of lung immunity.
Collapse
Affiliation(s)
- Carolina Lyon De Ana
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Virology, Immunology, & Microbiology, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Anukul T Shenoy
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department. of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Kimberly A Barker
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Virology, Immunology, & Microbiology, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Emad I Arafa
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Neelou S Etesami
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Virology, Immunology, & Microbiology, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Filiz T Korkmaz
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Alicia M Soucy
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Michael P Breen
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Virology, Immunology, & Microbiology, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Ian M C Martin
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Brian R Tilton
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Priyadharshini Devarajan
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Nicholas A Crossland
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA; Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Riley M F Pihl
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Flow Cytometry Core Facility, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Wesley N Goltry
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Anna C Belkina
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Flow Cytometry Core Facility, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Matthew R Jones
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Lee J Quinton
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Virology, Immunology, & Microbiology, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA; Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Virology, Immunology, & Microbiology, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Biochemistry & Cell Biology, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA.
| |
Collapse
|
11
|
Li N, Zhu J, Chen P, Bao C, Wang J, Abdelaal T, Chen D, Zhu S, Wang W, Mao J, Scicluna BP, Koning F, Li F, Lei L. High-dimensional analysis reveals an immune atlas and novel neutrophil clusters in the lungs of model animals with Actinobacillus pleuropneumoniae-induced pneumonia. Vet Res 2023; 54:76. [PMID: 37705063 PMCID: PMC10500746 DOI: 10.1186/s13567-023-01207-4] [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: 01/29/2023] [Accepted: 07/24/2023] [Indexed: 09/15/2023] Open
Abstract
Due to the increase in bacterial resistance, improving the anti-infectious immunity of the host is rapidly becoming a new strategy for the prevention and treatment of bacterial pneumonia. However, the specific lung immune responses and key immune cell subsets involved in bacterial infection are obscure. Actinobacillus pleuropneumoniae (APP) can cause porcine pleuropneumonia, a highly contagious respiratory disease that has caused severe economic losses in the swine industry. Here, using high-dimensional mass cytometry, the major immune cell repertoire in the lungs of mice with APP infection was profiled. Various phenotypically distinct neutrophil subsets and Ly-6C+ inflammatory monocytes/macrophages accumulated post-infection. Moreover, a linear differentiation trajectory from inactivated to activated to apoptotic neutrophils corresponded with the stages of uninfected, onset, and recovery of APP infection. CD14+ neutrophils, which mainly increased in number during the recovery stage of infection, were revealed to have a stronger ability to produce cytokines, especially IL-10 and IL-21, than their CD14- counterparts. Importantly, MHC-II+ neutrophils with antigen-presenting cell features were identified, and their numbers increased in the lung after APP infection. Similar results were further confirmed in the lungs of piglets infected with APP and Klebsiella pneumoniae infection by using a single-cell RNA-seq technique. Additionally, a correlation analysis between cluster composition and the infection process yielded a dynamic and temporally associated immune landscape where key immune clusters, including previously unrecognized ones, marked various stages of infection. Thus, these results reveal the characteristics of key neutrophil clusters and provide a detailed understanding of the immune response to bacterial pneumonia.
Collapse
Affiliation(s)
- Na Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Junhui Zhu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Peiru Chen
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Chuntong Bao
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jun Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Tamim Abdelaal
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, The Netherlands
- Department of Pattern Recognition and Bioinformatics Group, Delft University of Technology, Delft, The Netherlands
| | - Dexi Chen
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Sibo Zhu
- School of Life Sciences, Fudan University, Shanghai, China
| | - Wenjing Wang
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Jiangnan Mao
- School of Life Sciences, Fudan University, Shanghai, China
| | - Brendon P Scicluna
- Department of Applied Biomedical Science, Faculty of Health Sciences, Mater Dei Hospital, University of Malta, Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Frits Koning
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Fengyang Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Liancheng Lei
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China.
- College of Animal Science, Yangtze University, Jingzhou, Hubei, China.
| |
Collapse
|
12
|
Li M, Chen C, Wang X, Guo P, Feng H, Zhang X, Zhang W, Gu C, Zhu J, Wen G, Feng Y, Xiao L, Peng G, Rao VB, Tao P. T4 bacteriophage nanoparticles engineered through CRISPR provide a versatile platform for rapid development of flu mucosal vaccines. Antiviral Res 2023; 217:105688. [PMID: 37516153 DOI: 10.1016/j.antiviral.2023.105688] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 07/31/2023]
Abstract
Vaccines that trigger mucosal immune responses at the entry portals of pathogens are highly desired. Here, we showed that antigen-decorated nanoparticle generated through CRISPR engineering of T4 bacteriophage can serve as a universal platform for the rapid development of mucosal vaccines. Insertion of Flu viral M2e into phage T4 genome through fusion to Soc (Small Outer Capsid protein) generated a recombinant phage, and the Soc-M2e proteins self-assembled onto phage capsids to form 3M2e-T4 nanoparticles during propagation of T4 in E. coli. Intranasal administration of 3M2e-T4 nanoparticles maintains antigen persistence in the lungs, resulting in increased uptake and presentation by antigen-presenting cells. M2e-specific secretory IgA, effector (TEM), central (TCM), and tissue-resident memory CD4+ T cells (TRM) were efficiently induced in the local mucosal sites, which mediated protections against divergent influenza viruses. Our studies demonstrated the mechanisms of immune protection following 3M2e-T4 nanoparticles vaccination and provide a versatile T4 platform that can be customized to rapidly develop mucosal vaccines against future emerging epidemics.
Collapse
Affiliation(s)
- Mengling Li
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Cen Chen
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Xialin Wang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Pengju Guo
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Helong Feng
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, Hubei, 430070, China
| | - Xueqi Zhang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Wanpo Zhang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Changqin Gu
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Jingen Zhu
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC, 20064, USA
| | - Guoyuan Wen
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, Hubei, 430070, China
| | - Yaoyu Feng
- Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Lihua Xiao
- Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Guiqing Peng
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Venigalla B Rao
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC, 20064, USA
| | - Pan Tao
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China.
| |
Collapse
|
13
|
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
|
14
|
Abstract
Specialized subpopulations of CD4+ T cells survey major histocompatibility complex class II-peptide complexes to control phagosomal infections, help B cells, regulate tissue homeostasis and repair or perform immune regulation. Memory CD4+ T cells are positioned throughout the body and not only protect the tissues from reinfection and cancer, but also participate in allergy, autoimmunity, graft rejection and chronic inflammation. Here we provide updates on our understanding of the longevity, functional heterogeneity, differentiation, plasticity, migration and human immunodeficiency virus reservoirs as well as key technological advances that are facilitating the characterization of memory CD4+ T cell biology.
Collapse
Affiliation(s)
- Marco Künzli
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - David Masopust
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
15
|
Prior JT, Limbert VM, Horowitz RM, D'Souza SJ, Bachnak L, Godwin MS, Bauer DL, Harrell JE, Morici LA, Taylor JJ, McLachlan JB. Establishment of isotype-switched, antigen-specific B cells in multiple mucosal tissues using non-mucosal immunization. NPJ Vaccines 2023; 8:80. [PMID: 37258506 PMCID: PMC10231862 DOI: 10.1038/s41541-023-00677-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 05/18/2023] [Indexed: 06/02/2023] Open
Abstract
Although most pathogens infect the human body via mucosal surfaces, very few injectable vaccines can specifically target immune cells to these tissues where their effector functions would be most desirable. We have previously shown that certain adjuvants can program vaccine-specific helper T cells to migrate to the gut, even when the vaccine is delivered non-mucosally. It is not known whether this is true for antigen-specific B cell responses. Here we show that a single intradermal vaccination with the adjuvant double mutant heat-labile toxin (dmLT) induces a robust endogenous, vaccine-specific, isotype-switched B cell response. When the vaccine was intradermally boosted, we detected non-circulating vaccine-specific B cell responses in the lamina propria of the large intestines, Peyer's patches, and lungs. When compared to the TLR9 ligand adjuvant CpG, only dmLT was able to drive the establishment of isotype-switched resident B cells in these mucosal tissues, even when the dmLT-adjuvanted vaccine was administered non-mucosally. Further, we found that the transcription factor Batf3 was important for the full germinal center reaction, isotype switching, and Peyer's patch migration of these B cells. Collectively, these data indicate that specific adjuvants can promote mucosal homing and the establishment of activated, antigen-specific B cells in mucosal tissues, even when these adjuvants are delivered by a non-mucosal route. These findings could fundamentally change the way future vaccines are formulated and delivered.
Collapse
Affiliation(s)
- John T Prior
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Vanessa M Limbert
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Rebecca M Horowitz
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Shaina J D'Souza
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Louay Bachnak
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Matthew S Godwin
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - David L Bauer
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jaikin E Harrell
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Lisa A Morici
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Justin J Taylor
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - James B McLachlan
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA.
| |
Collapse
|
16
|
Iwanaga N, Devarajan P, Shenoy AT. Editorial: Adaptive immunity to respiratory pathogens. Front Immunol 2023; 14:1174178. [PMID: 36949940 PMCID: PMC10026996 DOI: 10.3389/fimmu.2023.1174178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Affiliation(s)
- Naoki Iwanaga
- Department of Respiratory Medicine, Nagasaki University Hospital, Nagasaki, Japan
| | - Priyadharshini Devarajan
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Anukul T. Shenoy
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| |
Collapse
|
17
|
Korkmaz FT, Traber KE. Innate immune responses in pneumonia. Pneumonia (Nathan) 2023; 15:4. [PMID: 36829255 PMCID: PMC9957695 DOI: 10.1186/s41479-023-00106-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 01/05/2023] [Indexed: 02/26/2023] Open
Abstract
The lungs are an immunologically unique environment; they are exposed to innumerable pathogens and particulate matter daily. Appropriate clearance of pathogens and response to pollutants is required to prevent overwhelming infection, while preventing tissue damage and maintaining efficient gas exchange. Broadly, the innate immune system is the collection of immediate, intrinsic immune responses to pathogen or tissue injury. In this review, we will examine the innate immune responses of the lung, with a particular focus on their role in pneumonia. We will discuss the anatomic barriers and antimicrobial proteins of the lung, pathogen and injury recognition, and the role of leukocytes (macrophages, neutrophils, and innate lymphocytes) and lung stromal cells in innate immunity. Throughout the review, we will focus on new findings in innate immunity as well as features that are unique to the lung.
Collapse
Affiliation(s)
- Filiz T Korkmaz
- Department of Medicine, Division of Immunology & Infectious Disease, University of Massachusetts, Worcester, MA, USA
- Pulmonary Center, Boston University School of Medicine, Boston, MA, USA
| | - Katrina E Traber
- Pulmonary Center, Boston University School of Medicine, Boston, MA, USA.
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
18
|
Korkmaz FT, Shenoy AT, Symer EM, Baird LA, Odom CV, Arafa EI, Dimbo EL, Na E, Molina-Arocho W, Brudner M, Standiford TJ, Mehta JL, Sawamura T, Jones MR, Mizgerd JP, Traber KE, Quinton LJ. Lectin-like oxidized low-density lipoprotein receptor 1 attenuates pneumonia-induced lung injury. JCI Insight 2022; 7:e149955. [PMID: 36264633 PMCID: PMC9746901 DOI: 10.1172/jci.insight.149955] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 10/18/2022] [Indexed: 01/12/2023] Open
Abstract
Identifying host factors that contribute to pneumonia incidence and severity are of utmost importance to guiding the development of more effective therapies. Lectin-like oxidized low-density lipoprotein receptor 1 (LOX-1, encoded by OLR1) is a scavenger receptor known to promote vascular injury and inflammation, but whether and how LOX-1 functions in the lung are unknown. Here, we provide evidence of substantial accumulation of LOX-1 in the lungs of patients with acute respiratory distress syndrome and in mice with pneumonia. Unlike previously described injurious contributions of LOX-1, we found that LOX-1 is uniquely protective in the pulmonary airspaces, limiting proteinaceous edema and inflammation. We also identified alveolar macrophages and recruited neutrophils as 2 prominent sites of LOX-1 expression in the lungs, whereby macrophages are capable of further induction during pneumonia and neutrophils exhibit a rapid, but heterogenous, elevation of LOX-1 in the infected lung. Blockade of LOX-1 led to dysregulated immune signaling in alveolar macrophages, marked by alterations in activation markers and a concomitant elevation of inflammatory gene networks. However, bone marrow chimeras also suggested a prominent role for neutrophils in LOX-1-mediated lung protection, further supported by LOX-1+ neutrophils exhibiting transcriptional changes consistent with reparative processes. Taken together, this work establishes LOX-1 as a tissue-protective factor in the lungs during pneumonia, possibly mediated by its influence on immune signaling in alveolar macrophages and LOX-1+ airspace neutrophils.
Collapse
Affiliation(s)
- Filiz T. Korkmaz
- Division of Immunology and Infectious Disease, Department of Medicine, UMass Chan Medical School, Worcester, Massachusetts, USA
| | | | | | | | | | | | | | | | | | - Matthew Brudner
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Theodore J. Standiford
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jawahar L. Mehta
- Department of Internal Medicine, College of Medicine, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| | - Tatsuya Sawamura
- Department of Molecular Pathophysiology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | | | - Joseph P. Mizgerd
- Pulmonary Center
- Department of Microbiology, and
- Department of Medicine and
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | | | - Lee J. Quinton
- Division of Immunology and Infectious Disease, Department of Medicine, UMass Chan Medical School, Worcester, Massachusetts, USA
- Pulmonary Center
- Department of Medicine and
| |
Collapse
|
19
|
Intranasal Vaccination with rePcrV Protects against Pseudomonas aeruginosa and Generates Lung Tissue-Resident Memory T Cells. J Immunol Res 2022; 2022:1403788. [DOI: 10.1155/2022/1403788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/09/2022] [Accepted: 11/11/2022] [Indexed: 11/28/2022] Open
Abstract
Tissue-resident memory T (TRM) cells are immune sentinels that bear a key role in the local immune system and rapidly respond to infection. Our previous studies showed that mucosal immunization via intranasal pathways was more effective than intramuscular route. However, the mechanism of enhanced protective immunity remains unclear. Here, we formulated a Pseudomonas aeruginosa vaccine composed of type III secretion protein PcrV from P. aeruginosa and curdlan adjuvant and then administered by the intranasal route. Flow cytometry and immunofluorescence staining showed that the ratio of CD44+CD62L-CD69+CD4+ TRM cells induced by this vaccine was significantly increased, and IL-17A production was notably enhanced. Further analysis revealed that vaccinated mice can protect against the P. aeruginosa challenge even after administration with FTY720 treatment. What is more, our results showed that CD4+ TRM might be involved in the recruitment of neutrophils and provided partial protection against Pseudomonas aeruginosa. Taken together, these data demonstrated that CD4+ TRM cells were elicited in lung tissues after immunization with rePcrV and contributed to protective immunity. Furthermore, it provided novel strategies for the development of vaccines for P. aeruginosa and other respiratory-targeted vaccines.
Collapse
|
20
|
Shenoy AT, De Ana CL, Barker KA, Arafa EI, Martin IM, Mizgerd JP, Belkina AC. CPHEN-011: Comprehensive phenotyping of murine lung resident lymphocytes after recovery from pneumococcal pneumonia. Cytometry A 2022; 101:892-902. [PMID: 34854229 PMCID: PMC9160214 DOI: 10.1002/cyto.a.24522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 01/27/2023]
Abstract
Recovery from pneumococcal (Spn) pneumonia induces development of tissue resident memory CD4+ TRM cells, BRM cells, and antibody secreting plasma cells in experienced lungs. These tissue resident lymphocytes confer protection against subsequent lethal challenge by serotype mismatched Spn (termed as heterotypic immunity). While traditional flow cytometry and gating strategies support premeditated identification of cells using a limited set of markers, discovery of novel tissue resident lymphocytes necessitates stable platforms that can handle larger sets of phenotypic markers and lends itself to unbiased clustering approaches. In this report, we leverage the power of full spectrum flow cytometry (FSFC) to develop a comprehensive panel of phenotypic markers that allows identification of multiple subsets of tissue resident lymphocytes in Spn-experienced murine lungs. Using Phenograph algorithm on this multidimensional data, we identify unforeseen heterogeneity in lung resident adaptive immune landscape which includes unexpected subsets of TRM and BRM cells. Further, using conventional gating strategy informed by our unsupervised clustering data, we confirm their presence exquisitely in Spn-experienced lungs as potentially relevant to heterotypic immunity and define CD73 as a highly expressed marker on TRM cells. Thus, our study emphasizes the utility of FSFC for confirmatory and discovery studies relating to tissue resident adaptive immunity.
Collapse
Affiliation(s)
- Anukul T. Shenoy
- Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Carolina Lyon De Ana
- Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA
- Dept. of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Kimberly A. Barker
- Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA
- Dept. of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Emad I. Arafa
- Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA
- Dept. of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Ian M.C. Martin
- Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Joseph P. Mizgerd
- Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA
- Dept. of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
- Dept. of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- Dept. of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Anna C. Belkina
- Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, 02118, USA
- Dept. of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
21
|
Rainard P, Gilbert FB, Germon P. Immune defenses of the mammary gland epithelium of dairy ruminants. Front Immunol 2022; 13:1031785. [PMID: 36341445 PMCID: PMC9634088 DOI: 10.3389/fimmu.2022.1031785] [Citation(s) in RCA: 13] [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: 08/30/2022] [Accepted: 10/03/2022] [Indexed: 11/17/2022] Open
Abstract
The epithelium of the mammary gland (MG) fulfills three major functions: nutrition of progeny, transfer of immunity from mother to newborn, and its own defense against infection. The defense function of the epithelium requires the cooperation of mammary epithelial cells (MECs) with intraepithelial leucocytes, macrophages, DCs, and resident lymphocytes. The MG is characterized by the secretion of a large amount of a nutrient liquid in which certain bacteria can proliferate and reach a considerable bacterial load, which has conditioned how the udder reacts against bacterial invasions. This review presents how the mammary epithelium perceives bacteria, and how it responds to the main bacterial genera associated with mastitis. MECs are able to detect the presence of actively multiplying bacteria in the lumen of the gland: they express pattern recognition receptors (PRRs) that recognize microbe-associated molecular patterns (MAMPs) released by the growing bacteria. Interactions with intraepithelial leucocytes fine-tune MECs responses. Following the onset of inflammation, new interactions are established with lymphocytes and neutrophils recruited from the blood. The mammary epithelium also identifies and responds to antigens, which supposes an antigen-presenting capacity. Its responses can be manipulated with drugs, plant extracts, probiotics, and immune modifiers, in order to increase its defense capacities or reduce the damage related to inflammation. Numerous studies have established that the mammary epithelium is a genuine effector of both innate and adaptive immunity. However, knowledge gaps remain and newly available tools offer the prospect of exciting research to unravel and exploit the multiple capacities of this particular epithelium.
Collapse
|
22
|
Rahimi RA, Cho JL, Jakubzick CV, Khader SA, Lambrecht BN, Lloyd CM, Molofsky AB, Talbot S, Bonham CA, Drake WP, Sperling AI, Singer BD. Advancing Lung Immunology Research: An Official American Thoracic Society Workshop Report. Am J Respir Cell Mol Biol 2022; 67:e1-18. [PMID: 35776495 PMCID: PMC9273224 DOI: 10.1165/rcmb.2022-0167st] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The mammalian airways and lungs are exposed to a myriad of inhaled particulate matter, allergens, and pathogens. The immune system plays an essential role in protecting the host from respiratory pathogens, but a dysregulated immune response during respiratory infection can impair pathogen clearance and lead to immunopathology. Furthermore, inappropriate immunity to inhaled antigens can lead to pulmonary diseases. A complex network of epithelial, neural, stromal, and immune cells has evolved to sense and respond to inhaled antigens, including the decision to promote tolerance versus a rapid, robust, and targeted immune response. Although there has been great progress in understanding the mechanisms governing immunity to respiratory pathogens and aeroantigens, we are only beginning to develop an integrated understanding of the cellular networks governing tissue immunity within the lungs and how it changes after inflammation and over the human life course. An integrated model of airway and lung immunity will be necessary to improve mucosal vaccine design as well as prevent and treat acute and chronic inflammatory pulmonary diseases. Given the importance of immunology in pulmonary research, the American Thoracic Society convened a working group to highlight central areas of investigation to advance the science of lung immunology and improve human health.
Collapse
|
23
|
Pathogenesis of pneumonia and acute lung injury. Clin Sci (Lond) 2022; 136:747-769. [PMID: 35621124 DOI: 10.1042/cs20210879] [Citation(s) in RCA: 101] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/29/2022] [Accepted: 05/09/2022] [Indexed: 12/15/2022]
Abstract
Pneumonia and its sequelae, acute lung injury, present unique challenges for pulmonary and critical care healthcare professionals, and these challenges have recently garnered global attention due to the ongoing Sars-CoV-2 pandemic. One limitation to translational investigation of acute lung injury, including its most severe manifestation (acute respiratory distress syndrome, ARDS) has been heterogeneity resulting from the clinical and physiologic diagnosis that represents a wide variety of etiologies. Recent efforts have improved our understanding and approach to heterogeneity by defining sub-phenotypes of ARDS although significant gaps in knowledge remain. Improving our mechanistic understanding of acute lung injury and its most common cause, infectious pneumonia, can advance our approach to precision targeted clinical interventions. Here, we review the pathogenesis of pneumonia and acute lung injury, including how respiratory infections and lung injury disrupt lung homoeostasis, and provide an overview of respiratory microbial pathogenesis, the lung microbiome, and interventions that have been demonstrated to improve outcomes-or not-in human clinical trials.
Collapse
|
24
|
Palmer CS, Kimmey JM. Neutrophil Recruitment in Pneumococcal Pneumonia. Front Cell Infect Microbiol 2022; 12:894644. [PMID: 35646729 PMCID: PMC9136017 DOI: 10.3389/fcimb.2022.894644] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/18/2022] [Indexed: 01/19/2023] Open
Abstract
Streptococcus pneumoniae (Spn) is the primary agent of community-acquired pneumonia. Neutrophils are innate immune cells that are essential for bacterial clearance during pneumococcal pneumonia but can also do harm to host tissue. Neutrophil migration in pneumococcal pneumonia is therefore a major determinant of host disease outcomes. During Spn infection, detection of the bacterium leads to an increase in proinflammatory signals and subsequent expression of integrins and ligands on both the neutrophil as well as endothelial and epithelial cells. These integrins and ligands mediate the tethering and migration of the neutrophil from the bloodstream to the site of infection. A gradient of host-derived and bacterial-derived chemoattractants contribute to targeted movement of neutrophils. During pneumococcal pneumonia, neutrophils are rapidly recruited to the pulmonary space, but studies show that some of the canonical neutrophil migratory machinery is dispensable. Investigation of neutrophil migration is necessary for us to understand the dynamics of pneumococcal infection. Here, we summarize what is known about the pathways that lead to migration of the neutrophil from the capillaries to the lung during pneumococcal infection.
Collapse
Affiliation(s)
| | - Jacqueline M. Kimmey
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, Santa Cruz, CA, United States
| |
Collapse
|
25
|
Kawasaki T, Ikegawa M, Kawai T. Antigen Presentation in the Lung. Front Immunol 2022; 13:860915. [PMID: 35615351 PMCID: PMC9124800 DOI: 10.3389/fimmu.2022.860915] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/15/2022] [Indexed: 12/28/2022] Open
Abstract
The lungs are constantly exposed to environmental and infectious agents such as dust, viruses, fungi, and bacteria that invade the lungs upon breathing. The lungs are equipped with an immune defense mechanism that involves a wide variety of immunological cells to eliminate these agents. Various types of dendritic cells (DCs) and macrophages (MACs) function as professional antigen-presenting cells (APCs) that engulf pathogens through endocytosis or phagocytosis and degrade proteins derived from them into peptide fragments. During this process, DCs and MACs present the peptides on their major histocompatibility complex class I (MHC-I) or MHC-II protein complex to naïve CD8+ or CD4+ T cells, respectively. In addition to these cells, recent evidence supports that antigen-specific effector and memory T cells are activated by other lung cells such as endothelial cells, epithelial cells, and monocytes through antigen presentation. In this review, we summarize the molecular mechanisms of antigen presentation by APCs in the lungs and their contribution to immune response.
Collapse
Affiliation(s)
| | | | - Taro Kawai
- *Correspondence: Takumi Kawasaki, ; Taro Kawai,
| |
Collapse
|
26
|
Zhao Z, Zhu H, Li Q, Liao W, Chen K, Yang M, Long D, He Z, Zhao M, Wu H, Lu Q. Skin CD4+ Trm cells distinguish acute cutaneous lupus erythematosus from localized discoid lupus erythematosus/subacute cutaneous lupus erythematosus and other skin diseases. J Autoimmun 2022; 128:102811. [DOI: 10.1016/j.jaut.2022.102811] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 12/30/2022]
|
27
|
Arafa EI, Shenoy AT, Barker KA, Etesami NS, Martin IM, Lyon De Ana C, Na E, Odom CV, Goltry WN, Korkmaz FT, Wooten AK, Belkina AC, Guillon A, Forsberg EC, Jones MR, Quinton LJ, Mizgerd JP. Recruitment and training of alveolar macrophages after pneumococcal pneumonia. JCI Insight 2022; 7:150239. [PMID: 35133985 PMCID: PMC8983128 DOI: 10.1172/jci.insight.150239] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 02/02/2022] [Indexed: 11/25/2022] Open
Abstract
Recovery from pneumococcal pneumonia remodels the pool of alveolar macrophages so that they exhibit new surface marker profiles, transcriptomes, metabolomes, and responses to infection. Mechanisms mediating alveolar macrophage phenotypes after pneumococcal pneumonia have not been delineated. IFN-γ and its receptor on alveolar macrophages were essential for certain, but not all, aspects of the remodeled alveolar macrophage phenotype. IFN-γ was produced by CD4+ T cells plus other cells, and CD4+ cell depletion did not prevent alveolar macrophage remodeling. In mice infected or recovering from pneumococcus, monocytes were recruited to the lungs, and the monocyte-derived macrophages developed characteristics of alveolar macrophages. CCR2 mediated the early monocyte recruitment but was not essential to the development of the remodeled alveolar macrophage phenotype. Lineage tracing demonstrated that recovery from pneumococcal pneumonias converted the pool of alveolar macrophages from being primarily of embryonic origin to being primarily of adult hematopoietic stem cell origin. Alveolar macrophages of either origin demonstrated similar remodeled phenotypes, suggesting that ontogeny did not dictate phenotype. Our data reveal that the remodeled alveolar macrophage phenotype in lungs recovered from pneumococcal pneumonia results from a combination of new recruitment plus training of both the original cells and the new recruits.
Collapse
Affiliation(s)
- Emad I Arafa
- Pulmonary Center, Boston University School of Medicine, Boston, United States of America
| | - Anukul T Shenoy
- Pulmonary Center, Boston University School of Medicine, Boston, United States of America
| | - Kimberly A Barker
- Pulmonary Center, Boston University School of Medicine, Boston, United States of America
| | - Neelou S Etesami
- Department of Microbiology, Boston University School of Medicine, Boston, United States of America
| | - Ian Mc Martin
- Pulmonary Center, Boston University School of Medicine, Boston, United States of America
| | - Carolina Lyon De Ana
- Pulmonary Center, Boston University School of Medicine, Boston, United States of America
| | - Elim Na
- Pulmonary Center, Boston University School of Medicine, Boston, United States of America
| | - Christine V Odom
- Pulmonary Center, Boston University School of Medicine, Boston, United States of America
| | - Wesley N Goltry
- Pulmonary Center, Boston University School of Medicine, Boston, United States of America
| | - Filiz T Korkmaz
- Pulmonary Center, Boston University School of Medicine, Boston, United States of America
| | - Alicia K Wooten
- Pulmonary Center, Boston University School of Medicine, Boston, United States of America
| | - Anna C Belkina
- Pulmonary Center, Boston University School of Medicine, Boston, United States of America
| | - Antoine Guillon
- CHRU of Tours, service de Médecine Intensive Réanimation, University of Tours, Tours, France
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, University of California Santa Cruz, Santa Cruz, United States of America
| | - Matthew R Jones
- Pulmonary Center, Boston University School of Medicine, Boston, United States of America
| | - Lee J Quinton
- Pulmonary Center, Boston University School of Medicine, Boston, United States of America
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University School of Medicine, Boston, United States of America
| |
Collapse
|
28
|
Zheng MZM, Wakim LM. Tissue resident memory T cells in the respiratory tract. Mucosal Immunol 2022; 15:379-388. [PMID: 34671115 PMCID: PMC8526531 DOI: 10.1038/s41385-021-00461-z] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/27/2021] [Accepted: 10/01/2021] [Indexed: 02/04/2023]
Abstract
Owing to their capacity to rapidly spread across the population, airborne pathogens represent a significant risk to global health. Indeed, several of the past major global pandemics have been instigated by respiratory pathogens. A greater understanding of the immune cells tasked with protecting the airways from infection will allow for the development of strategies that curb the spread and impact of these airborne diseases. A specific subset of memory T-cell resident in both the upper and lower respiratory tract, termed tissue-resident memory (Trm), have been shown to play an instrumental role in local immune responses against a wide breadth of both viral and bacterial infections. In this review, we discuss factors that influence respiratory tract Trm development, longevity, and immune surveillance and explore vaccination regimes that harness these cells, such approaches represent exciting new strategies that may be utilized to tackle the next global pandemic.
Collapse
Affiliation(s)
- Ming Z. M. Zheng
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| | - Linda M. Wakim
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000 Australia
| |
Collapse
|
29
|
Tissue-resident immunity in the lung: a first-line defense at the environmental interface. Semin Immunopathol 2022; 44:827-854. [PMID: 36305904 PMCID: PMC9614767 DOI: 10.1007/s00281-022-00964-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 09/08/2022] [Indexed: 12/15/2022]
Abstract
The lung is a vital organ that incessantly faces external environmental challenges. Its homeostasis and unimpeded vital function are ensured by the respiratory epithelium working hand in hand with an intricate fine-tuned tissue-resident immune cell network. Lung tissue-resident immune cells span across the innate and adaptive immunity and protect from infectious agents but can also prove to be pathogenic if dysregulated. Here, we review the innate and adaptive immune cell subtypes comprising lung-resident immunity and discuss their ontogeny and role in distinct respiratory diseases. An improved understanding of the role of lung-resident immunity and how its function is dysregulated under pathological conditions can shed light on the pathogenesis of respiratory diseases.
Collapse
|
30
|
Song D, Yan F, Fu H, Li L, Hao J, Zhu Z, Ye L, Zhang Y, Jin M, Dai L, Fang H, Song Z, Wu D, Wang X. A cellular census of human peripheral immune cells identifies novel cell states in lung diseases. Clin Transl Med 2021; 11:e579. [PMID: 34841705 PMCID: PMC8611783 DOI: 10.1002/ctm2.579] [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: 03/27/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 12/19/2022] Open
Abstract
Increasing evidence supports a central role of the immune system in lung diseases. Understanding how immunological alterations between lung diseases provide opportunities for immunotherapy. Exhausted T cells play a key role of immune suppression in lung cancer and chronic obstructive pulmonary disease was proved in our previous study. The present study aims to furthermore define molecular landscapes and heterogeneity of systemic immune cell target proteomic and transcriptomic profiles and interactions between circulating immune cells and lung residential cells in various lung diseases. We firstly measured target proteomic profiles of circulating immune cells from healthy volunteers and patients with stable pneumonia, stable asthma, acute asthma, acute exacerbation of chronic obstructive pulmonary disease, chronic obstructive pulmonary disease and lung cancer, using single-cell analysis by cytometry by time-of-flight with 42 antibodies. The nine immune cells landscape was mapped among those respiratory system diseases, including CD4+ T cells, CD8+ T cells, dendritic cells, B cells, eosinophil, γδT cells, monocytes, neutrophil and natural killer cells. The double-negative T cells and exhausted CD4+ central memory T cells subset were identified in patients with acute pneumonia. This T subset expressed higher levels of T-cell immunoglobulin and mucin domain-containing protein 3 (Tim3) and T-cell immunoreceptor with Ig and ITIM domains (TIGIT) in patients with acute pneumonia and stable pneumonia. Biological processes and pathways of immune cells including immune response activation, regulation of cell cycle and pathways in cancer in peripheral blood immune cells were defined by bulk RNA sequencing (RNA-seq). The heterogeneity among immune cells including CD4+ , CD8+ T cells and NK T cells by single immune cell RNA-seq with significant difference was found by single-cell sequencing. The effect of interstitial telocytes on the immune cell types and immune function was finally studied and the expressions of CD8a and chemokine C-C motif receptor 7 (CCR7) were increased significantly in co-cultured groups. Our data indicate that proteomic and transcriptomic profiles and heterogeneity of circulating immune cells provides new insights for understanding new molecular mechanisms of immune cell function, interaction and modulation as a source to identify and develop biomarkers and targets for lung diseases.
Collapse
Affiliation(s)
- Dongli Song
- Zhongshan HospitalDepartment of Pulmonary and Critical Care MedicineInstitute for Clinical ScienceShanghai Medical UniversityFudan UniversityShanghaiChina
| | - Furong Yan
- Zhongshan HospitalDepartment of Pulmonary and Critical Care MedicineInstitute for Clinical ScienceShanghai Medical UniversityFudan UniversityShanghaiChina
| | - Huirong Fu
- Zhongshan HospitalDepartment of Pulmonary and Critical Care MedicineInstitute for Clinical ScienceShanghai Medical UniversityFudan UniversityShanghaiChina
| | - Liyang Li
- Zhongshan HospitalDepartment of Pulmonary and Critical Care MedicineInstitute for Clinical ScienceShanghai Medical UniversityFudan UniversityShanghaiChina
| | - Jie Hao
- Zhongshan HospitalDepartment of Pulmonary and Critical Care MedicineInstitute for Clinical ScienceShanghai Medical UniversityFudan UniversityShanghaiChina
| | - Zhenhua Zhu
- Zhongshan HospitalDepartment of Pulmonary and Critical Care MedicineInstitute for Clinical ScienceShanghai Medical UniversityFudan UniversityShanghaiChina
| | - Ling Ye
- Zhongshan HospitalDepartment of Pulmonary and Critical Care MedicineInstitute for Clinical ScienceShanghai Medical UniversityFudan UniversityShanghaiChina
| | - Yong Zhang
- Zhongshan HospitalDepartment of Pulmonary and Critical Care MedicineInstitute for Clinical ScienceShanghai Medical UniversityFudan UniversityShanghaiChina
| | - Meiling Jin
- Zhongshan HospitalDepartment of Pulmonary and Critical Care MedicineInstitute for Clinical ScienceShanghai Medical UniversityFudan UniversityShanghaiChina
| | - Lihua Dai
- Department of EmergencyShidong Hospital of Yangpu DistrictShanghaiChina
| | - Hao Fang
- Department of AnesthesiologyZhongshan HospitalShanghai Medical UniversityFudan UniversityShanghaiChina
| | - Zhenju Song
- Department of EmergencyZhongshan HospitalShanghai Medical UniversityFudan UniversityShanghaiChina
| | - Duojiao Wu
- Zhongshan HospitalDepartment of Pulmonary and Critical Care MedicineInstitute for Clinical ScienceShanghai Medical UniversityFudan UniversityShanghaiChina
- Shanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
- Jinshan Hospital Centre for Tumour Diagnosis and TherapyShanghai Medical UniversityFudan UniversityShanghaiChina
| | - Xiangdong Wang
- Zhongshan HospitalDepartment of Pulmonary and Critical Care MedicineInstitute for Clinical ScienceShanghai Medical UniversityFudan UniversityShanghaiChina
- Shanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
- Jinshan Hospital Centre for Tumour Diagnosis and TherapyShanghai Medical UniversityFudan UniversityShanghaiChina
| |
Collapse
|
31
|
Yuan R, Yu J, Jiao Z, Li J, Wu F, Yan R, Huang X, Chen C. The Roles of Tissue-Resident Memory T Cells in Lung Diseases. Front Immunol 2021; 12:710375. [PMID: 34707601 PMCID: PMC8542931 DOI: 10.3389/fimmu.2021.710375] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 09/27/2021] [Indexed: 12/25/2022] Open
Abstract
The unique environment of the lungs is protected by complex immune interactions. Human lung tissue-resident memory T cells (TRM) have been shown to position at the pathogen entry points and play an essential role in fighting against viral and bacterial pathogens at the frontline through direct mechanisms and also by orchestrating the adaptive immune system through crosstalk. Recent evidence suggests that TRM cells also play a vital part in slowing down carcinogenesis and preventing the spread of solid tumors. Less beneficially, lung TRM cells can promote pathologic inflammation, causing chronic airway inflammatory changes such as asthma and fibrosis. TRM cells from infiltrating recipient T cells may also mediate allograft immunopathology, hence lung damage in patients after lung transplantations. Several therapeutic strategies targeting TRM cells have been developed. This review will summarize recent advances in understanding the establishment and maintenance of TRM cells in the lung, describe their roles in different lung diseases, and discuss how the TRM cells may guide future immunotherapies targeting infectious diseases, cancers and pathologic immune responses.
Collapse
Affiliation(s)
- Rui Yuan
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Jiang Yu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ziqiao Jiao
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Jinfei Li
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Fang Wu
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Rongkai Yan
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Xiaojie Huang
- Department Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Chen Chen
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China.,Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
32
|
Shenoy AT, Lyon De Ana C, Arafa EI, Salwig I, Barker KA, Korkmaz FT, Ramanujan A, Etesami NS, Soucy AM, Martin IMC, Tilton BR, Hinds A, Goltry WN, Kathuria H, Braun T, Jones MR, Quinton LJ, Belkina AC, Mizgerd JP. Antigen presentation by lung epithelial cells directs CD4 + T RM cell function and regulates barrier immunity. Nat Commun 2021; 12:5834. [PMID: 34611166 PMCID: PMC8492657 DOI: 10.1038/s41467-021-26045-w] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 08/31/2021] [Indexed: 12/17/2022] Open
Abstract
Barrier tissues are populated by functionally plastic CD4+ resident memory T (TRM) cells. Whether the barrier epithelium regulates CD4+ TRM cell locations, plasticity and activities remains unclear. Here we report that lung epithelial cells, including distinct surfactant protein C (SPC)lowMHChigh epithelial cells, function as anatomically-segregated and temporally-dynamic antigen presenting cells. In vivo ablation of lung epithelial MHC-II results in altered localization of CD4+ TRM cells. Recurrent encounters with cognate antigen in the absence of epithelial MHC-II leads CD4+ TRM cells to co-express several classically antagonistic lineage-defining transcription factors, changes their cytokine profiles, and results in dysregulated barrier immunity. In addition, lung epithelial MHC-II is needed for surface expression of PD-L1, which engages its ligand PD-1 to constrain lung CD4+ TRM cell phenotypes. Thus, we establish epithelial antigen presentation as a critical regulator of CD4+ TRM cell function and identify epithelial-CD4+ TRM cell immune interactions as core elements of barrier immunity.
Collapse
Affiliation(s)
- Anukul T Shenoy
- Pulmonary Center, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Carolina Lyon De Ana
- Pulmonary Center, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Microbiology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Emad I Arafa
- Pulmonary Center, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Isabelle Salwig
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Kimberly A Barker
- Pulmonary Center, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Microbiology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Filiz T Korkmaz
- Pulmonary Center, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Aditya Ramanujan
- Pulmonary Center, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Neelou S Etesami
- Pulmonary Center, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Microbiology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Alicia M Soucy
- Pulmonary Center, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Ian M C Martin
- Pulmonary Center, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Brian R Tilton
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Anne Hinds
- Pulmonary Center, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Wesley N Goltry
- Pulmonary Center, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Hasmeena Kathuria
- Pulmonary Center, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Microbiology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Matthew R Jones
- Pulmonary Center, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Lee J Quinton
- Pulmonary Center, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Microbiology, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Anna C Belkina
- Pulmonary Center, Boston University School of Medicine, Boston, MA, 02118, USA
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University School of Medicine, Boston, MA, 02118, USA.
- Department of Microbiology, Boston University School of Medicine, Boston, MA, 02118, USA.
- Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA.
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA.
| |
Collapse
|
33
|
Iwanaga N, Chen K, Yang H, Lu S, Hoffmann JP, Wanek A, McCombs JE, Song K, Rangel-Moreno J, Norton EB, Kolls JK. Vaccine-driven lung TRM cells provide immunity against Klebsiella via fibroblast IL-17R signaling. Sci Immunol 2021; 6:eabf1198. [PMID: 34516780 DOI: 10.1126/sciimmunol.abf1198] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Naoki Iwanaga
- Departments of Pediatrics and Medicine, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Kong Chen
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Haoran Yang
- Departments of Pediatrics and Medicine, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Shiping Lu
- Departments of Pediatrics and Medicine, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Joseph P Hoffmann
- Departments of Pediatrics and Medicine, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Alanna Wanek
- Departments of Pediatrics and Medicine, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Janet E McCombs
- Departments of Pediatrics and Medicine, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Kejing Song
- Departments of Pediatrics and Medicine, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | - Elizabeth B Norton
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jay K Kolls
- Departments of Pediatrics and Medicine, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
34
|
Mac Aogáin M, Baker JM, Dickson RP. On Bugs and Blowholes: Why Is Aspiration the Rule, Not the Exception? Am J Respir Crit Care Med 2021; 203:1049-1051. [PMID: 33596387 PMCID: PMC8314891 DOI: 10.1164/rccm.202011-4257ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Micheál Mac Aogáin
- Department of Biochemistry St. James's Hospital Dublin, Ireland.,School of Medicine Trinity College Dublin Dublin, Ireland
| | - Jennifer M Baker
- Department of Internal Medicine University of Michigan Medical School Ann Arbor, Michigan.,Department of Microbiology and Immunology University of Michigan Medical School Ann Arbor, Michigan and
| | - Robert P Dickson
- Department of Internal Medicine University of Michigan Medical School Ann Arbor, Michigan.,Department of Microbiology and Immunology University of Michigan Medical School Ann Arbor, Michigan and.,Michigan Center for Integrative Research in Critical Care Ann Arbor, Michigan
| |
Collapse
|
35
|
Clegg J, Soldaini E, McLoughlin RM, Rittenhouse S, Bagnoli F, Phogat S. Staphylococcus aureus Vaccine Research and Development: The Past, Present and Future, Including Novel Therapeutic Strategies. Front Immunol 2021; 12:705360. [PMID: 34305945 PMCID: PMC8294057 DOI: 10.3389/fimmu.2021.705360] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/22/2021] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus is one of the most important human pathogens worldwide. Its high antibiotic resistance profile reinforces the need for new interventions like vaccines in addition to new antibiotics. Vaccine development efforts against S. aureus have failed so far however, the findings from these human clinical and non-clinical studies provide potential insight for such failures. Currently, research is focusing on identifying novel vaccine formulations able to elicit potent humoral and cellular immune responses. Translational science studies are attempting to discover correlates of protection using animal models as well as in vitro and ex vivo models assessing efficacy of vaccine candidates. Several new vaccine candidates are being tested in human clinical trials in a variety of target populations. In addition to vaccines, bacteriophages, monoclonal antibodies, centyrins and new classes of antibiotics are being developed. Some of these have been tested in humans with encouraging results. The complexity of the diseases and the range of the target populations affected by this pathogen will require a multipronged approach using different interventions, which will be discussed in this review.
Collapse
Affiliation(s)
- Jonah Clegg
- GSK, Siena, Italy
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | - Rachel M. McLoughlin
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | | |
Collapse
|
36
|
Carlier FM, de Fays C, Pilette C. Epithelial Barrier Dysfunction in Chronic Respiratory Diseases. Front Physiol 2021; 12:691227. [PMID: 34248677 PMCID: PMC8264588 DOI: 10.3389/fphys.2021.691227] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022] Open
Abstract
Mucosal surfaces are lined by epithelial cells, which provide a complex and adaptive module that ensures first-line defense against external toxics, irritants, antigens, and pathogens. The underlying mechanisms of host protection encompass multiple physical, chemical, and immune pathways. In the lung, inhaled agents continually challenge the airway epithelial barrier, which is altered in chronic diseases such as chronic obstructive pulmonary disease, asthma, cystic fibrosis, or pulmonary fibrosis. In this review, we describe the epithelial barrier abnormalities that are observed in such disorders and summarize current knowledge on the mechanisms driving impaired barrier function, which could represent targets of future therapeutic approaches.
Collapse
Affiliation(s)
- François M. Carlier
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
- Department of Pneumology and Lung Transplant, Centre Hospitalier Universitaire UCL Namur, Yvoir, Belgium
| | - Charlotte de Fays
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Charles Pilette
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
- Department of Pneumology, Cliniques universitaires St-Luc, Brussels, Belgium
| |
Collapse
|
37
|
Walkowski W, Bassett J, Bhalla M, Pfeifer BA, Ghanem ENB. Intranasal Vaccine Delivery Technology for Respiratory Tract Disease Application with a Special Emphasis on Pneumococcal Disease. Vaccines (Basel) 2021; 9:vaccines9060589. [PMID: 34199398 PMCID: PMC8230341 DOI: 10.3390/vaccines9060589] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/17/2021] [Accepted: 05/22/2021] [Indexed: 12/17/2022] Open
Abstract
This mini-review will cover recent trends in intranasal (IN) vaccine delivery as it relates to applications for respiratory tract diseases. The logic and rationale for IN vaccine delivery will be compared to methods and applications accompanying this particular administration route. In addition, we will focus extended discussion on the potential role of IN vaccination in the context of respiratory tract diseases, with a special emphasis on pneumococcal disease. Here, elements of this disease, including its prevalence and impact upon the elderly population, will be viewed from the standpoint of improving health outcomes through vaccine design and delivery technology and how IN administration can play a role in such efforts.
Collapse
Affiliation(s)
- William Walkowski
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA; (W.W.); (J.B.); (B.A.P.)
| | - Justin Bassett
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA; (W.W.); (J.B.); (B.A.P.)
| | - Manmeet Bhalla
- Department of Microbiology and Immunology, University at Buffalo, The State University of New York, Buffalo, NY 14203, USA;
| | - Blaine A. Pfeifer
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA; (W.W.); (J.B.); (B.A.P.)
| | - Elsa N. Bou Ghanem
- Department of Microbiology and Immunology, University at Buffalo, The State University of New York, Buffalo, NY 14203, USA;
- Correspondence:
| |
Collapse
|
38
|
Barker KA, Etesami NS, Shenoy AT, Arafa EI, Lyon de Ana C, Smith NM, Martin IM, Goltry WN, Barron AM, Browning JL, Kathuria H, Belkina AC, Guillon A, Zhong X, Crossland NA, Jones MR, Quinton LJ, Mizgerd JP. Lung-resident memory B cells protect against bacterial pneumonia. J Clin Invest 2021; 131:e141810. [PMID: 34060477 DOI: 10.1172/jci141810] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 04/14/2021] [Indexed: 12/22/2022] Open
Abstract
Lung-resident memory B cells (BRM cells) are elicited after influenza infections of mice, but connections to other pathogens and hosts - as well as their functional significance - have yet to be determined. We postulate that BRM cells are core components of lung immunity. To test this, we examined whether lung BRM cells are elicited by the respiratory pathogen pneumococcus, are present in humans, and are important in pneumonia defense. Lungs of mice that had recovered from pneumococcal infections did not contain organized tertiary lymphoid organs, but did have plasma cells and noncirculating memory B cells. The latter expressed distinctive surface markers (including CD69, PD-L2, CD80, and CD73) and were poised to secrete antibodies upon stimulation. Human lungs also contained B cells with a resident memory phenotype. In mice recovered from pneumococcal pneumonia, depletion of PD-L2+ B cells, including lung BRM cells, diminished bacterial clearance and the level of pneumococcus-reactive antibodies in the lung. These data define lung BRM cells as a common feature of pathogen-experienced lungs and provide direct evidence of a role for these cells in pulmonary antibacterial immunity.
Collapse
Affiliation(s)
| | | | | | | | | | - Nicole Ms Smith
- Pulmonary Center.,Department of Pathology and Laboratory Medicine, and
| | | | | | | | | | | | - Anna C Belkina
- Pulmonary Center.,Department of Pathology and Laboratory Medicine, and.,Flow Cytometry Core Facility, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Antoine Guillon
- Pulmonary Center.,Centre Hospitalier Régional Universitaire de (CHRU) de Tours, Service de Médecine Intensive Réanimation, INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, University of Tours, Tours, France
| | | | | | | | - Lee J Quinton
- Pulmonary Center.,Department of Microbiology.,Department of Medicine.,Department of Pathology and Laboratory Medicine, and
| | - Joseph P Mizgerd
- Pulmonary Center.,Department of Microbiology.,Department of Medicine.,Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
39
|
LeibundGut-Landmann S. Tissue-Resident Memory T Cells in Antifungal Immunity. Front Immunol 2021; 12:693055. [PMID: 34113356 PMCID: PMC8185520 DOI: 10.3389/fimmu.2021.693055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/10/2021] [Indexed: 12/26/2022] Open
Abstract
Fungi are an integral part of the mammalian microbiota colonizing most if not all mucosal surfaces and the skin. Maintaining stable colonization on these surfaces is critical for preventing fungal dysbiosis and infection, which in some cases can lead to life threatening consequences. The epithelial barriers are protected by T cells and additional controlling immune mechanisms. Noncirculating memory T cells that reside stably in barrier tissues play an important role for host protection from commensals and recurrent pathogens due to their fast response and local activity, which provides them a strategic advantage. So far, only a few specific examples of tissue resident memory T cells (TRMs) that act against fungi have been reported. This review provides an overview of the characteristics and functional attributes of TRMs that have been established based on human and mouse studies with various microbes. It highlights what is currently known about fungi specific TRMs mediating immunosurveillance, how they have been targeted in preclinical vaccination approaches and how they can promote immunopathology, if not controlled. A better appreciation of the host protective and damaging roles of TRMs might accelerate the development of novel tissue specific preventive strategies against fungal infections and fungi-driven immunopathologies.
Collapse
Affiliation(s)
- Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
40
|
Nur Husna SM, Tan HTT, Md Shukri N, Mohd Ashari NS, Wong KK. Nasal Epithelial Barrier Integrity and Tight Junctions Disruption in Allergic Rhinitis: Overview and Pathogenic Insights. Front Immunol 2021; 12:663626. [PMID: 34093555 PMCID: PMC8176953 DOI: 10.3389/fimmu.2021.663626] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/22/2021] [Indexed: 12/20/2022] Open
Abstract
Allergic rhinitis (AR) is a common disorder affecting up to 40% of the population worldwide and it usually persists throughout life. Nasal epithelial barrier constitutes the first line of defense against invasion of harmful pathogens or aeroallergens. Cell junctions comprising of tight junctions (TJs), adherens junctions, desmosomes and hemidesmosomes form the nasal epithelial barrier. Impairment of TJ molecules plays causative roles in the pathogenesis of AR. In this review, we describe and discuss the components of TJs and their disruption leading to development of AR, as well as regulation of TJs expression by epigenetic changes, neuro-immune interaction, epithelial-derived cytokines (thymic stromal lymphopoietin, IL-25 and IL-33), T helper 2 (Th2) cytokines (IL-4, IL-5, IL-6 and IL-13) and innate lymphoid cells. These growing evidence support the development of novel therapeutic approaches to restore nasal epithelial TJs expression in AR patients.
Collapse
Affiliation(s)
- Siti Muhamad Nur Husna
- Department of Immunology, School of Medical Sciences Malaysia, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Hern-Tze Tina Tan
- Department of Immunology, School of Medical Sciences Malaysia, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Norasnieda Md Shukri
- Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia.,Department of Otorhinolaryngology, Head and Neck Surgery, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Noor Suryani Mohd Ashari
- Department of Immunology, School of Medical Sciences Malaysia, Universiti Sains Malaysia, Kubang Kerian, Malaysia.,Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Kah Keng Wong
- Department of Immunology, School of Medical Sciences Malaysia, Universiti Sains Malaysia, Kubang Kerian, Malaysia.,Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
41
|
Shenoy AT, Mizgerd JP. Seedy CD8+ T RM cells in aging lungs drive susceptibility to pneumonia and sequelae. Cell Mol Immunol 2021; 18:787-789. [PMID: 33420355 PMCID: PMC7791330 DOI: 10.1038/s41423-020-00629-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 12/11/2020] [Indexed: 11/15/2022] Open
Affiliation(s)
- Anukul T Shenoy
- Pulmonary Center, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University School of Medicine, Boston, MA, 02118, USA.
- Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA.
- Department of Microbiology, Boston University School of Medicine, Boston, MA, 02118, USA.
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA.
| |
Collapse
|
42
|
Dijkman K, Aguilo N, Boot C, Hofman SO, Sombroek CC, Vervenne RA, Kocken CH, Marinova D, Thole J, Rodríguez E, Vierboom MP, Haanstra KG, Puentes E, Martin C, Verreck FA. Pulmonary MTBVAC vaccination induces immune signatures previously correlated with prevention of tuberculosis infection. Cell Rep Med 2021; 2:100187. [PMID: 33521701 PMCID: PMC7817873 DOI: 10.1016/j.xcrm.2020.100187] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 10/23/2020] [Accepted: 12/17/2020] [Indexed: 11/29/2022]
Abstract
To fight tuberculosis, better vaccination strategies are needed. Live attenuated Mycobacterium tuberculosis-derived vaccine, MTBVAC, is a promising candidate in the pipeline, proven to be safe and immunogenic in humans so far. Independent studies have shown that pulmonary mucosal delivery of Bacillus Calmette-Guérin (BCG), the only tuberculosis (TB) vaccine available today, confers superior protection over standard intradermal immunization. Here we demonstrate that mucosal MTBVAC is well tolerated, eliciting polyfunctional T helper type 17 cells, interleukin-10, and immunoglobulins in the airway and yielding a broader antigenic profile than BCG in rhesus macaques. Beyond our previous work, we show that local immunoglobulins, induced by MTBVAC and BCG, bind to M. tuberculosis and enhance pathogen uptake. Furthermore, after pulmonary vaccination, but not M. tuberculosis infection, local T cells expressed high levels of mucosal homing and tissue residency markers. Our data show that pulmonary MTBVAC administration has the potential to enhance its efficacy and justifies further exploration of mucosal vaccination strategies in preclinical efficacy studies.
Collapse
Affiliation(s)
- Karin Dijkman
- Biomedical Primate Research Centre (BPRC), Rijswijk, the Netherlands
| | - Nacho Aguilo
- Department of Microbiology, Faculty of Medicine, IIS Aragon, University of Zaragoza, Zaragoza, Spain
- CIBERES, Instituto de Salud Carlos III, Madrid, Spain
| | - Charelle Boot
- Biomedical Primate Research Centre (BPRC), Rijswijk, the Netherlands
| | - Sam O. Hofman
- Biomedical Primate Research Centre (BPRC), Rijswijk, the Netherlands
| | | | | | | | - Dessislava Marinova
- Department of Microbiology, Faculty of Medicine, IIS Aragon, University of Zaragoza, Zaragoza, Spain
- CIBERES, Instituto de Salud Carlos III, Madrid, Spain
| | - Jelle Thole
- TuBerculosis Vaccine Initiative (TBVI), Lelystad, the Netherlands
| | | | | | | | | | - Carlos Martin
- Department of Microbiology, Faculty of Medicine, IIS Aragon, University of Zaragoza, Zaragoza, Spain
- CIBERES, Instituto de Salud Carlos III, Madrid, Spain
| | | |
Collapse
|
43
|
Abstract
The community of cells lining our airways plays a collaborative role in the preservation of immune homeostasis in the lung and provides protection from the pathogens and pollutants in the air we breathe. In addition to its structural attributes that provide effective mucociliary clearance of the lower airspace, the airway epithelium is an immunologically active barrier surface that senses changes in the airway environment and interacts with resident and recruited immune cells. Single-cell RNA-sequencing is illuminating the cellular heterogeneity that exists in the airway wall and has identified novel cell populations with unique molecular signatures, trajectories of differentiation and diverse functions in health and disease. In this Review, we discuss how our view of the airway epithelial landscape has evolved with the advent of transcriptomic approaches to cellular phenotyping, with a focus on epithelial interactions with the local neuronal and immune systems.
Collapse
|
44
|
IL-17 mediates protective immunity against nasal infection with Bordetella pertussis by mobilizing neutrophils, especially Siglec-F + neutrophils. Mucosal Immunol 2021; 14:1183-1202. [PMID: 33976385 PMCID: PMC8379078 DOI: 10.1038/s41385-021-00407-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 03/26/2021] [Accepted: 04/15/2021] [Indexed: 02/04/2023]
Abstract
Understanding the mechanism of protective immunity in the nasal mucosae is central to the design of more effective vaccines that prevent nasal infection and transmission of Bordetella pertussis. We found significant infiltration of IL-17-secreting CD4+ tissue-resident memory T (TRM) cells and Siglec-F+ neutrophils into the nasal tissue during primary infection with B. pertussis. Il17A-/- mice had significantly higher bacterial load in the nasal mucosae, associated with significantly reduced infiltration of Siglec-F+ neutrophils. Re-infected convalescent mice rapidly cleared B. pertussis from the nasal cavity and this was associated with local expansion of IL-17-producing CD4+ TRM cells. Depletion of CD4 T cells from the nasal tissue during primary infection or after re-challenge of convalescent mice significantly delayed clearance of bacteria from the nasal mucosae. Protection was lost in Il17A-/- mice and this was associated with significantly less infiltration of Siglec-F+ neutrophils and antimicrobial peptide (AMP) production. Finally, depletion of neutrophils reduced the clearance of B. pertussis following re-challenge of convalescent mice. Our findings demonstrate that IL-17 plays a critical role in natural and acquired immunity to B. pertussis in the nasal mucosae and this effect is mediated by mobilizing neutrophils, especially Siglec-F+ neutrophils, which have high neutrophil extracellular trap (NET) activity.
Collapse
|
45
|
Clegg J, Soldaini E, Bagnoli F, McLoughlin RM. Targeting Skin-Resident Memory T Cells via Vaccination to Combat Staphylococcus aureus Infections. Trends Immunol 2020; 42:6-17. [PMID: 33309137 DOI: 10.1016/j.it.2020.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023]
Abstract
Tissue-resident memory T cells are important in adaptive immunity against many infections, rendering these cells attractive potential targets in vaccine development. Genetic and experimental evidence highlights the importance of cellular immunity in protection from Staphylococcus aureus skin infections, yet skin-resident memory T cells are, thus far, an untested component of immunity during such infections. Novel methods of generating and sampling vaccine-induced skin memory T cells are paralleled by discoveries of global, skin-wide immunosurveillance. We propose skin-resident memory CD4+ T cells as a potential missing link in the search for correlates of protection during S. aureus infections. A better appreciation of their phenotypes and functions could accelerate the development of preventive vaccines against this highly virulent and antibiotic-resistant pathogen.
Collapse
Affiliation(s)
- Jonah Clegg
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; GlaxoSmithKline, Siena, Italy
| | | | | | - Rachel M McLoughlin
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
46
|
Xiong G, Xie X, Wang Q, Zhang Y, Ge Y, Lin W, Li M. Immune cell infiltration and related core genes expression characteristics in eosinophilic and non-eosinophilic chronic rhinosinusitis with nasal polyps. Exp Ther Med 2020; 20:180. [PMID: 33101470 PMCID: PMC7579783 DOI: 10.3892/etm.2020.9310] [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: 02/27/2020] [Accepted: 09/04/2020] [Indexed: 12/31/2022] Open
Abstract
Chronic rhinosinusitis with nasal polyps (CRSwNP) refers to chronic inflammation of the sinonasal mucosa. It can either be eosinophilic (ECRSwNP) or non-eosinophilic (non-ECRSwNP). However, immune cell infiltration in the microenvironment and pathogenesis of ECRSwNP and non-ECRSwNP are still unclear. The aim of the present study was to assess the immune cell infiltration and molecular mechanisms of ECRSwNP and non-ECRSwNP. In the present study, 22 immune cell types in ECRSwNP and non-ECRSwNP were investigated by CIBERSORT based on transcriptome data. The core gene related pathophysiology of CRSwNP was analyzed using Weighted Gene Correlation Network Analysis according to the phenotype of the infiltrated eosinophils and nasal polyps (NP). A total of four types of immune cells (mast cells, activated dendritic cells, M2 macrophages and activated natural killer cells) were demonstrated to have a direct and indirect correlation with eosinophilic infiltration in ECRSwNP. M1 macrophages and activated CD4+ memory T cells were correlated with major immune cell types in non-ECRSwNP. NP could affect the expression of ‘olfactory receptor activity’ and ‘channel activity’ genes to impair the olfactory signaling pathway and neuroactive ligand receptor pathway. ‘Cell adhesion molecule binding’, ‘cytokine receptor binding’ and ‘glucocorticoid receptor binding’ were significantly enriched in ECRSwNP, whereas epithelial cell injury, autophagy and the mTOR pathway (hsa04140 and hsa04150) may serve an important role in the pathogenesis of non-ECRSwNP. There were significantly different immune cell infiltration and related core genes expression characteristics between ECRSwNP and non-ECRSwNP. The results of the present study provide an improved basis for elucidation of the mechanism and treatment of CRSwNP.
Collapse
Affiliation(s)
- Gaoyun Xiong
- Department of Otolaryngology, Head and Neck Surgery, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Xiaoxing Xie
- Department of Otolaryngology, Head and Neck Surgery, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Qingliang Wang
- Department of Otolaryngology, Head and Neck Surgery, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Yanyan Zhang
- Department of Otolaryngology, Head and Neck Surgery, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Yanping Ge
- Department of Otolaryngology, Head and Neck Surgery, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Wei Lin
- Department of Otolaryngology, Head and Neck Surgery, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Mingqian Li
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| |
Collapse
|
47
|
Xing Z, Afkhami S, Bavananthasivam J, Fritz DK, D'Agostino MR, Vaseghi-Shanjani M, Yao Y, Jeyanathan M. Innate immune memory of tissue-resident macrophages and trained innate immunity: Re-vamping vaccine concept and strategies. J Leukoc Biol 2020; 108:825-834. [PMID: 32125045 DOI: 10.1002/jlb.4mr0220-446r] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 02/03/2020] [Accepted: 02/09/2020] [Indexed: 02/06/2023] Open
Abstract
In the past few years, our understanding of immunological memory has evolved remarkably due to a growing body of new knowledge in innate immune memory and immunity. Immunological memory now encompasses both innate and adaptive immune memory. The hypo-reactive and hyper-reactive types of innate immune memory lead to a suppressed and enhanced innate immune protective outcome, respectively. The latter is also named trained innate immunity (TII). The emerging information on innate immune memory has not only shed new light on the mechanisms of host defense but is also revolutionizing our long-held view of vaccination and vaccine strategies. Our current review will examine recent progress and knowledge gaps in innate immune memory with a focus on tissue-resident Mϕs, particularly lung Mϕs, and their relationship to local antimicrobial innate immunity. We will also discuss the impact of innate immune memory and TII on our understanding of vaccine concept and strategies and the significance of respiratory mucosal route of vaccination against respiratory pathogens.
Collapse
Affiliation(s)
- Zhou Xing
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Sam Afkhami
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jegarubee Bavananthasivam
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Dominik K Fritz
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Michael R D'Agostino
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Maryam Vaseghi-Shanjani
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Yushi Yao
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.,Current affiliation: Department of Immunology, Zhejiang University, Zhejiang, China
| | - Mangalakumari Jeyanathan
- McMaster Immunology Research Centre, Hamilton, Ontario, Canada.,M. G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada.,Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
48
|
Guillon A, Arafa EI, Barker KA, Belkina AC, Martin I, Shenoy AT, Wooten AK, Lyon De Ana C, Dai A, Labadorf A, Hernandez Escalante J, Dooms H, Blasco H, Traber KE, Jones MR, Quinton LJ, Mizgerd JP. Pneumonia recovery reprograms the alveolar macrophage pool. JCI Insight 2020; 5:133042. [PMID: 31990682 PMCID: PMC7101156 DOI: 10.1172/jci.insight.133042] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 01/22/2020] [Indexed: 12/21/2022] Open
Abstract
Community-acquired pneumonia is a widespread disease with significant morbidity and mortality. Alveolar macrophages are tissue-resident lung cells that play a crucial role in innate immunity against bacteria that cause pneumonia. We hypothesized that alveolar macrophages display adaptive characteristics after resolution of bacterial pneumonia. We studied mice 1 to 6 months after self-limiting lung infections with Streptococcus pneumoniae, the most common cause of bacterial pneumonia. Alveolar macrophages, but not other myeloid cells, recovered from the lung showed long-term modifications of their surface marker phenotype. The remodeling of alveolar macrophages was (a) long-lasting (still observed 6 months after infection), (b) regionally localized (observed only in the affected lobe after lobar pneumonia), and (c) associated with macrophage-dependent enhanced protection against another pneumococcal serotype. Metabolomic and transcriptomic profiling revealed that alveolar macrophages of mice that recovered from pneumonia had new baseline activities and altered responses to infection that better resembled those of adult humans. The enhanced lung protection after mild and self-limiting bacterial respiratory infections includes a profound remodeling of the alveolar macrophage pool that is long-lasting; compartmentalized; and manifest across surface receptors, metabolites, and both resting and stimulated transcriptomes.
Collapse
Affiliation(s)
- Antoine Guillon
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
- CHRU of Tours, service de Médecine Intensive Réanimation, INSERM, Centre d’Etude des Pathologies Respiratoires (CEPR), UMR 1100, University of Tours, Tours, France
| | - Emad I. Arafa
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Medicine
| | - Kimberly A. Barker
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Microbiology
| | - Anna C. Belkina
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Pathology and Laboratory Medicine, and
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Ian Martin
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Anukul T. Shenoy
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Alicia K. Wooten
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Medicine
| | - Carolina Lyon De Ana
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Microbiology
| | - Anqi Dai
- Bioinformatics Nexus, Boston University, Boston, Massachusetts, USA
| | - Adam Labadorf
- Bioinformatics Nexus, Boston University, Boston, Massachusetts, USA
| | | | - Hans Dooms
- Department of Medicine
- Department of Microbiology
| | - Hélène Blasco
- CHRU of Tours, Medical Pharmacology Department, Inserm U1253, University of Tours, Tours, France
| | - Katrina E. Traber
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Medicine
| | - Matthew R. Jones
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Medicine
| | - Lee J. Quinton
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Medicine
- Department of Microbiology
- Department of Pathology and Laboratory Medicine, and
| | - Joseph P. Mizgerd
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Medicine
- Department of Microbiology
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
49
|
Larsen SB, Cowley CJ, Fuchs E. Epithelial cells: liaisons of immunity. Curr Opin Immunol 2019; 62:45-53. [PMID: 31874430 DOI: 10.1016/j.coi.2019.11.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/13/2019] [Indexed: 01/12/2023]
Abstract
The surface and lining tissues of our body are exposed to the external environment, and as such these epithelial tissues must form structural barriers able to defend against microbes, environmental toxins, and mechanical stress. Their cells are equipped to detect a diverse array of surface perturbations, and then launch signaling relays to the immune system. The aim of these liaisons is to coordinate the requisite immune cell response needed to preserve and/or restore barrier integrity and defend the host. It has been recently appreciated that epithelial cells learn from these experiences. Following inflammatory exposure, long-lived stem cells within the tissue retain an epigenetic memory that endows them with heightened responsiveness to subsequent encounters with stress. Here, we review the recent literature on how epithelial cells sense signals from microbes, allergens, and injury at the tissue surface, and transmit this information to immune cells, while embedding a memory of the experience within their chromatin.
Collapse
Affiliation(s)
- Samantha B Larsen
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA; New York University School of Medicine, Neuroscience Institute, New York, NY 10016, USA
| | - Christopher J Cowley
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Elaine Fuchs
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|