1
|
Alladina J, Medoff BD, Cho JL. Innate Immunity and Asthma Exacerbations: Insights From Human Models. Immunol Rev 2025; 330:e70016. [PMID: 40087882 PMCID: PMC11922041 DOI: 10.1111/imr.70016] [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: 01/13/2025] [Revised: 02/14/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
Asthma is a common chronic respiratory disease characterized by the presence of airway inflammation, airway hyperresponsiveness, and mucus hypersecretion. Repeated asthma exacerbations can lead to progressive airway remodeling and irreversible airflow obstruction. Thus, understanding and preventing asthma exacerbations are of paramount importance. Although multiple endotypes exist, asthma is most often driven by type 2 airway inflammation. New therapies that target specific type 2 mediators have been shown to reduce the frequency of asthma exacerbations but are incompletely effective in a significant number of asthmatics. Furthermore, it remains unknown whether current treatments lead to sustained changes in the airway or if targeting additional pathways may be necessary to achieve asthma remission. Activation of innate immunity is the initial event in the inflammatory sequence that occurs during an asthma exacerbation. However, there continue to be critical gaps in our understanding of the innate immune response to asthma exacerbating factors. In this review, we summarize the current understanding of the role of innate immunity in asthma exacerbations and the methods used to study them. We also identify potential novel therapeutic targets for asthma and future areas for investigation.
Collapse
Affiliation(s)
- Jehan Alladina
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Benjamin D. Medoff
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Josalyn L. Cho
- Division of Pulmonary, Critical Care and Occupational Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
2
|
Seydoux E, Fytianos K, Garnier CV, Rothen-Rutishauser B, Blank F. Targeting Immune Cells. J Aerosol Med Pulm Drug Deliv 2024; 37:328-337. [PMID: 39625807 DOI: 10.1089/jamp.2024.63954.es] [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] [Indexed: 12/17/2024] Open
Abstract
The respiratory tract with its vast surface area and very thin air-blood tissue barrier presents an extremely large interface for potential interaction with xenobiotics such as inhaled pathogens or medicaments. To protect its large and vulnerable surface, the lung is populated with several different types of immune cells. Pulmonary epithelial cells, macrophages and dendritic cells are key players in shaping the innate and adaptive immune response. Due to their localization, they represent a frontline of cell populations that are among the first to come in contact with inhaled xenobiotics. Furthermore, depending on the lung compartment they populate, these cells show a large variety in morphology, phenotype, and function. These unique characteristics make those cell populations ideal targets for specific immunomodulators that are designed for inhalation. Depending on cell population or lung compartment targeting, a specific immune response may be triggered or modulated. The purpose of a potent carrier for pulmonary immunomodulation is, first, to efficiently target a specific immunocompetent cell and, second, to affect its role in generating an immune response. Immunomodulation may occur at different levels of immune cell-antigen interaction, i.e. antigen uptake, trafficking, processing and presentation. Inhalation of nanosized carriers for drugs or vaccines shows great potential for both prophylactic and therapeutic approaches in order to modulate immune responses locally or systemically, due to the specific deposition and targeting properties of nanoparticles. Immune responses triggered by nanosized particles may be either immunostimulatory or immunosuppressive and depending on the specific purpose, stimulation or suppression may either be desired or unwanted. Meticulous analysis of immunomodulatory potential, pharmacologic and toxicologic testing of inhalable nanocarriers is required in order to find novel and optimal approaches for prophylaxis and therapy of pulmonary diseases. The design and characterization of such nanoparticles requires well-coordinated interdisciplinary research among engineers, biologists and clinicians.
Collapse
Affiliation(s)
- Emilie Seydoux
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Kleanthis Fytianos
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | | | | | - Fabian Blank
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
3
|
Hargitai R, Parráková L, Szatmári T, Monfort-Lanzas P, Galbiati V, Audouze K, Jornod F, Staal YCM, Burla S, Chary A, Gutleb AC, Lumniczky K, Vandebriel RJ, Gostner JM. Chemical respiratory sensitization-Current status of mechanistic understanding, knowledge gaps and possible identification methods of sensitizers. FRONTIERS IN TOXICOLOGY 2024; 6:1331803. [PMID: 39135743 PMCID: PMC11317441 DOI: 10.3389/ftox.2024.1331803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 05/27/2024] [Indexed: 08/15/2024] Open
Abstract
Respiratory sensitization is a complex immunological process eventually leading to hypersensitivity following re-exposure to the chemical. A frequent consequence is occupational asthma, which may occur after long latency periods. Although chemical-induced respiratory hypersensitivity has been known for decades, there are currently no comprehensive and validated approaches available for the prospective identification of chemicals that induce respiratory sensitization, while the expectations of new approach methodologies (NAMs) are high. A great hope is that due to a better understanding of the molecular key events, new methods can be developed now. However, this is a big challenge due to the different chemical classes to which respiratory sensitizers belong, as well as because of the complexity of the response and the late manifestation of symptoms. In this review article, the current information on respiratory sensitization related processes is summarized by introducing it in the available adverse outcome pathway (AOP) concept. Potentially useful models for prediction are discussed. Knowledge gaps and gaps of regulatory concern are identified.
Collapse
Affiliation(s)
- Rita Hargitai
- Unit of Radiation Medicine, Department of Radiobiology and Radiohygiene, National Centre for Public Health and Pharmacy (NCPHP), Budapest, Hungary
| | - Lucia Parráková
- Biochemical Immunotoxicology Group, Institute of Medical Biochemistry, Medical University of Innsbruck (MUI), Innsbruck, Austria
| | - Tünde Szatmári
- Unit of Radiation Medicine, Department of Radiobiology and Radiohygiene, National Centre for Public Health and Pharmacy (NCPHP), Budapest, Hungary
| | - Pablo Monfort-Lanzas
- Biochemical Immunotoxicology Group, Institute of Medical Biochemistry, Medical University of Innsbruck (MUI), Innsbruck, Austria
- Institute of Bioinformatics, Medical University of Innsbruck (MUI), Innsbruck, Austria
| | - Valentina Galbiati
- Laboratory of Toxicology, Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università Degli Studi di Milano (UNIMI), Milano, Italy
| | | | | | - Yvonne C. M. Staal
- Centre for Health Protection, National Institute of Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Sabina Burla
- Luxembourg Institute of Science and Technology (LIST), Belvaux, Luxembourg
| | - Aline Chary
- Luxembourg Institute of Science and Technology (LIST), Belvaux, Luxembourg
| | - Arno C. Gutleb
- Luxembourg Institute of Science and Technology (LIST), Belvaux, Luxembourg
| | - Katalin Lumniczky
- Unit of Radiation Medicine, Department of Radiobiology and Radiohygiene, National Centre for Public Health and Pharmacy (NCPHP), Budapest, Hungary
| | - Rob J. Vandebriel
- Centre for Health Protection, National Institute of Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Johanna M. Gostner
- Biochemical Immunotoxicology Group, Institute of Medical Biochemistry, Medical University of Innsbruck (MUI), Innsbruck, Austria
| |
Collapse
|
4
|
Ma Y, Jiang T, Zhu X, Xu Y, Wan K, Zhang T, Xie M. Efferocytosis in dendritic cells: an overlooked immunoregulatory process. Front Immunol 2024; 15:1415573. [PMID: 38835772 PMCID: PMC11148234 DOI: 10.3389/fimmu.2024.1415573] [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: 04/10/2024] [Accepted: 05/09/2024] [Indexed: 06/06/2024] Open
Abstract
Efferocytosis, the process of engulfing and removing apoptotic cells, plays an essential role in preserving tissue health and averting undue inflammation. While macrophages are primarily known for this task, dendritic cells (DCs) also play a significant role. This review delves into the unique contributions of various DC subsets to efferocytosis, highlighting the distinctions in how DCs and macrophages recognize and handle apoptotic cells. It further explores how efferocytosis influences DC maturation, thereby affecting immune tolerance. This underscores the pivotal role of DCs in orchestrating immune responses and sustaining immune equilibrium, providing new insights into their function in immune regulation.
Collapse
Affiliation(s)
- Yanyan Ma
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tangxing Jiang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xun Zhu
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yizhou Xu
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ke Wan
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tingxuan Zhang
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Miaorong Xie
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Wang Y, Huang X, Luo G, Xu Y, Deng X, Lin Y, Wang Z, Zhou S, Wang S, Chen H, Tao T, He L, Yang L, Yang L, Chen Y, Jin Z, He C, Han Z, Zhang X. The aging lung: microenvironment, mechanisms, and diseases. Front Immunol 2024; 15:1383503. [PMID: 38756780 PMCID: PMC11096524 DOI: 10.3389/fimmu.2024.1383503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/16/2024] [Indexed: 05/18/2024] Open
Abstract
With the development of global social economy and the deepening of the aging population, diseases related to aging have received increasing attention. The pathogenesis of many respiratory diseases remains unclear, and lung aging is an independent risk factor for respiratory diseases. The aging mechanism of the lung may be involved in the occurrence and development of respiratory diseases. Aging-induced immune, oxidative stress, inflammation, and telomere changes can directly induce and promote the occurrence and development of lung aging. Meanwhile, the occurrence of lung aging also further aggravates the immune stress and inflammatory response of respiratory diseases; the two mutually affect each other and promote the development of respiratory diseases. Explaining the mechanism and treatment direction of these respiratory diseases from the perspective of lung aging will be a new idea and research field. This review summarizes the changes in pulmonary microenvironment, metabolic mechanisms, and the progression of respiratory diseases associated with aging.
Collapse
Affiliation(s)
- Yanmei Wang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Institute of Traditional Chinese Medicine of Sichuan Academy of Chinese Medicine Sciences (Sichuan Second Hospital of T.C.M), Chengdu, China
| | - Xuewen Huang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guofeng Luo
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunying Xu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiqian Deng
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yumeng Lin
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhanzhan Wang
- Department of Respiratory and Critical Care Medicine, The First People’s Hospital of Lianyungang, Lianyungang, China
| | - Shuwei Zhou
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Siyu Wang
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Haoran Chen
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tao Tao
- Institute of Traditional Chinese Medicine of Sichuan Academy of Chinese Medicine Sciences (Sichuan Second Hospital of T.C.M), Chengdu, China
| | - Lei He
- Institute of Traditional Chinese Medicine of Sichuan Academy of Chinese Medicine Sciences (Sichuan Second Hospital of T.C.M), Chengdu, China
| | - Luchuan Yang
- Institute of Traditional Chinese Medicine of Sichuan Academy of Chinese Medicine Sciences (Sichuan Second Hospital of T.C.M), Chengdu, China
| | - Li Yang
- Institute of Traditional Chinese Medicine of Sichuan Academy of Chinese Medicine Sciences (Sichuan Second Hospital of T.C.M), Chengdu, China
| | - Yutong Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zi Jin
- Department of Anesthesiology and Pain Rehabilitation, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Chengshi He
- Department of Respiratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhongyu Han
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaohong Zhang
- Department of Emergency Medicine Center, Sichuan Province People’s Hospital University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
6
|
Alotaibi N, Aldahlawi A, Zaher K, Basingab F, Alrahimi J. Optimizing the generation of mature bone marrow-derived dendritic cells in vitro: a factorial study design. J Genet Eng Biotechnol 2023; 21:144. [PMID: 38017248 PMCID: PMC10684437 DOI: 10.1186/s43141-023-00597-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/09/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Factorial design is a simple, yet elegant method to investigate the effect of multiple factors and their interaction on a specific response simultaneously. Hence, this type of study design reaches the best optimization conditions of a process. Although the interaction between the variables is widely prevalent in cell culture procedures, factorial design per se is infrequently utilized in improving cell culture output. Therefore, we aim to optimize the experimental conditions for generating mature bone marrow-derived dendritic cells (BMDCs). Two different variables were investigated, including the concentrations of the inducing factors and the starting density of the bone marrow mononuclear cells. In the current study, we utilized the design of experiments (DoE), a statistical approach, to systematically assess the impact of factors with varying levels on cell culture outcomes. Herein, we apply a two-factor, two-level (22) factorial experiment resulting in four conditions that are run in triplicate. The two variables investigated here are cytokines combinations with two levels, granulocyte-macrophage colony-stimulating factor (GM-CSF) alone or with interleukin-4 (IL4). The other parameter is cell density with two different concentrations, 2 × 106 and 4 × 106 cells/mL. Then, we measured cell viability using the trypan blue exclusion method, and a flow cytometer was used to detect the BMDCs expressing the markers FITC-CD80, CD86, CD83, and CD14. BMDC marker expression levels were calculated using arbitrary units (AU) of the mean fluorescence intensity (MFI). RESULTS The current study showed that the highest total viable cells and cells yield obtained were in cell group seeded at 2 × 106 cells/mL and treated with GM-CSF and IL-4. Importantly, the expression of the co-stimulatory molecules CD83 and CD80/CD86 were statistically significant for cell density of 2 × 106 cells/mL (P < 0.01, two-way ANOVA). Bone marrow mononuclear cells seeded at 4 × 106 in the presence of the cytokine mix less efficiently differentiated and matured into BMDCs. Statistical analysis via two-way ANOVA revealed an interaction between cell density and cytokine combinations. CONCLUSION The analysis of this study indicates a substantial interaction between cytokines combinations and cell densities on BMDC maturation. However, higher cell density is not associated with optimizing DC maturation. Notably, applying DoE in bioprocess designs increases experimental efficacy and reliability while minimizing experiments, time, and process costs.
Collapse
Affiliation(s)
- Najla Alotaibi
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
- Immunology Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.
- College of Health, Oregon State University, Corvallis, OR, USA.
| | - Alia Aldahlawi
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Immunology Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Kawther Zaher
- Immunology Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fatemah Basingab
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Immunology Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jehan Alrahimi
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Immunology Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
7
|
Probst HC, Stoitzner P, Amon L, Backer RA, Brand A, Chen J, Clausen BE, Dieckmann S, Dudziak D, Heger L, Hodapp K, Hornsteiner F, Hovav AH, Jacobi L, Ji X, Kamenjarin N, Lahl K, Lahmar I, Lakus J, Lehmann CHK, Ortner D, Picard M, Roberti MP, Rossnagel L, Saba Y, Schalla C, Schlitzer A, Schraml BU, Schütze K, Seichter A, Seré K, Seretis A, Sopper S, Strandt H, Sykora MM, Theobald H, Tripp CH, Zitvogel L. Guidelines for DC preparation and flow cytometry analysis of mouse nonlymphoid tissues. Eur J Immunol 2023; 53:e2249819. [PMID: 36512638 DOI: 10.1002/eji.202249819] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 12/15/2022]
Abstract
This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs and various nonlymphoid tissues. DC are sentinels of the immune system present in almost every mammalian organ. Since they represent a rare cell population, DC need to be extracted from organs with protocols that are specifically developed for each tissue. This article provides detailed protocols for the preparation of single-cell suspensions from various mouse nonlymphoid tissues, including skin, intestine, lung, kidney, mammary glands, oral mucosa and transplantable tumors. Furthermore, our guidelines include comprehensive protocols for multiplex flow cytometry analysis of DC subsets and feature top tricks for their proper discrimination from other myeloid cells. With this collection, we provide guidelines for in-depth analysis of DC subsets that will advance our understanding of their respective roles in healthy and diseased tissues. While all protocols were written by experienced scientists who routinely use them in their work, this article was also peer-reviewed by leading experts and approved by all coauthors, making it an essential resource for basic and clinical DC immunologists.
Collapse
Affiliation(s)
- Hans Christian Probst
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Patrizia Stoitzner
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Ronald A Backer
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Anna Brand
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Jianzhou Chen
- Gustave Roussy Cancer Campus (GRCC), U1015 INSERM, University Paris Saclay, Villejuif, France
| | - Björn E Clausen
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Sophie Dieckmann
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
- Medical Immunology Campus Erlangen (MICE), D-91054, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Germany
- Friedrich-Alexander University (FAU), Erlangen-Nürnberg, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Katrin Hodapp
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Florian Hornsteiner
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Avi-Hai Hovav
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Lukas Jacobi
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Xingqi Ji
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, 82152, Planegg-Martinsried, Germany
- Institute for Cardiovascular Physiology and Pathophysiology, Biomedical Center, Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Nadine Kamenjarin
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Katharina Lahl
- Section for Experimental and Translational Immunology, Institute for Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, 2800, Denmark
- Immunology Section, Lund University, Lund, 221 84, Sweden
| | - Imran Lahmar
- Gustave Roussy Cancer Campus (GRCC), U1015 INSERM, University Paris Saclay, Villejuif, France
| | - Jelena Lakus
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
- Medical Immunology Campus Erlangen (MICE), D-91054, Erlangen, Germany
| | - Daniela Ortner
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Marion Picard
- Gustave Roussy Cancer Campus (GRCC), U1015 INSERM, University Paris Saclay, Villejuif, France
| | - Maria Paula Roberti
- Gustave Roussy Cancer Campus (GRCC), U1015 INSERM, University Paris Saclay, Villejuif, France
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD), Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lukas Rossnagel
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Yasmin Saba
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Carmen Schalla
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Andreas Schlitzer
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Barbara U Schraml
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, 82152, Planegg-Martinsried, Germany
- Institute for Cardiovascular Physiology and Pathophysiology, Biomedical Center, Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Kristian Schütze
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Anna Seichter
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Kristin Seré
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Athanasios Seretis
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Sieghart Sopper
- Internal Medicine V, Hematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
- Tyrolean Cancer Research Center, Innsbruck, Austria
| | - Helen Strandt
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Martina M Sykora
- Internal Medicine V, Hematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
- Tyrolean Cancer Research Center, Innsbruck, Austria
| | - Hannah Theobald
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Christoph H Tripp
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus (GRCC), U1015 INSERM, University Paris Saclay, Villejuif, France
| |
Collapse
|
8
|
Chen K, Gong W, Huang J, Yoshimura T, Ming Wang J. Developmental and homeostatic signaling transmitted by the G-protein coupled receptor FPR2. Int Immunopharmacol 2023; 118:110052. [PMID: 37003185 PMCID: PMC10149111 DOI: 10.1016/j.intimp.2023.110052] [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: 12/29/2022] [Revised: 03/02/2023] [Accepted: 03/15/2023] [Indexed: 04/03/2023]
Abstract
Formyl peptide receptor 2 (FPR2) and its mouse counterpart Fpr2 are the members of the G protein-coupled receptor (GPCR) family. FPR2 is the only member of the FPRs that interacts with ligands from different sources. FPR2 is expressed in myeloid cells as well as epithelial cells, endothelial cells, neurons, and hepatocytes. During the past years, some unusual properties of FPR2 have attracted intense attention because FPR2 appears to possess dual functions by activating or inhibiting intracellular signal pathways based on the nature, concentration of the ligands, and the temporal and spatial settings of the microenvironment in vivo, the cell types it interacts with. Therefore, FPR2 controls an abundant array of developmental and homeostatic signaling cascades, in addition to its "classical" capacity to mediate the migration of hematopoietic and non-hematopoietic cells including malignant cells. In this review, we summarize recent development in FPR2 research, particularly in its role in diseases, therefore helping to establish FPR2 as a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Keqiang Chen
- Laboratory of Cancer Innovation, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA.
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Jiaqiang Huang
- Laboratory of Cancer Innovation, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA; College of Life Sciences, Beijing Jiaotong University, Beijing, PR China
| | - Teizo Yoshimura
- Laboratory of Cancer Innovation, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| | - Ji Ming Wang
- Laboratory of Cancer Innovation, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| |
Collapse
|
9
|
Trivedi A, Reed HO. The lymphatic vasculature in lung function and respiratory disease. Front Med (Lausanne) 2023; 10:1118583. [PMID: 36999077 PMCID: PMC10043242 DOI: 10.3389/fmed.2023.1118583] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/23/2023] [Indexed: 03/18/2023] Open
Abstract
The lymphatic vasculature maintains tissue homeostasis via fluid drainage in the form of lymph and immune surveillance due to migration of leukocytes through the lymphatics to the draining lymph nodes. Lymphatic endothelial cells (LECs) form the lymphatic vessels and lymph node sinuses and are key players in shaping immune responses and tolerance. In the healthy lung, the vast majority of lymphatic vessels are found along the bronchovascular structures, in the interlobular septa, and in the subpleural space. Previous studies in both mice and humans have shown that the lymphatics are necessary for lung function from the neonatal period through adulthood. Furthermore, changes in the lymphatic vasculature are observed in nearly all respiratory diseases in which they have been analyzed. Recent work has pointed to a causative role for lymphatic dysfunction in the initiation and progression of lung disease, indicating that these vessels may be active players in pathologic processes in the lung. However, the mechanisms by which defects in lung lymphatic function are pathogenic are understudied, leaving many unanswered questions. A more comprehensive understanding of the mechanistic role of morphological, functional, and molecular changes in the lung lymphatic endothelium in respiratory diseases is a promising area of research that is likely to lead to novel therapeutic targets. In this review, we will discuss our current knowledge of the structure and function of the lung lymphatics and the role of these vessels in lung homeostasis and respiratory disease.
Collapse
Affiliation(s)
- Anjali Trivedi
- Weill Cornell Medical Center, New York, NY, United States
| | - Hasina Outtz Reed
- Weill Cornell Medical Center, New York, NY, United States
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, United States
- *Correspondence: Hasina Outtz Reed,
| |
Collapse
|
10
|
Conti BJ, Santiago KB, Cardoso EO, Conte FL, Golim MA, Cruz MT, Sforcin JM. Effect of propolis on Th2 and Th17 cells: interplay with EtxB- and LPS-treated dendritic cells. Braz J Med Biol Res 2023; 56:e12659. [PMID: 37075347 PMCID: PMC10125804 DOI: 10.1590/1414-431x2023e12659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/07/2023] [Indexed: 04/21/2023] Open
Abstract
Dendritic cells (DCs) are antigen-presenting cells that drive the differentiation of T CD4+ cells into different profiles according to the nature of the antigen or immunomodulator. Propolis is a resinous product made by bees that has numerous pharmacological properties, including an immunomodulatory action. To assess whether propolis can modulate the activation of CD4+ T cells by stimulating DCs with heat-labile enterotoxin B subunit (EtxB) or lipopolysaccharide (LPS), we aimed to elucidate the mechanisms affected by propolis in the differential activation of T lymphocytes. Cell viability, lymphocyte proliferation, gene expression (GATA-3 and RORc), and cytokine production (interleukin (IL)-4 and IL-17A) were analyzed. Propolis, EtxB, and LPS induced a higher lymphoproliferation compared with the control. Propolis induced GATA-3 expression and, in combination with EtxB, maintained the baseline levels. Propolis alone or in combination with LPS inhibited RORc expression. EtxB alone and in combination with propolis increased IL-4 production. Propolis in combination with LPS prevented LPS-induced IL-17A production. These results opened perspectives for the study of biological events that may be favored by propolis by promoting Th2 activation or helping in the treatment of inflammatory conditions mediated by Th17 cells.
Collapse
Affiliation(s)
- B J Conti
- Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - K B Santiago
- Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - E O Cardoso
- Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - F L Conte
- Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - M A Golim
- Hemocentro de Botucatu, Faculdade de Medicina, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - M T Cruz
- Faculty of Pharmacy, Center for Neurosciences and Cellular Biology, University of Coimbra, Coimbra, Portugal
| | - J M Sforcin
- Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brasil
| |
Collapse
|
11
|
Furlong-Silva J, Cook PC. Fungal-mediated lung allergic airway disease: The critical role of macrophages and dendritic cells. PLoS Pathog 2022; 18:e1010608. [PMID: 35834490 PMCID: PMC9282651 DOI: 10.1371/journal.ppat.1010608] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Fungi are abundant in the environment, causing our lungs to be constantly exposed to a diverse range of species. While the majority of these are cleared effectively in healthy individuals, constant exposure to spores (especially Aspergillus spp.) can lead to the development of allergic inflammation that underpins and worsen diseases such as asthma. Despite this, the precise mechanisms that underpin the development of fungal allergic disease are poorly understood. Innate immune cells, such as macrophages (MΦs) and dendritic cells (DCs), have been shown to be critical for mediating allergic inflammation to a range of different allergens. This review will focus on the crucial role of MΦ and DCs in mediating antifungal immunity, evaluating how these immune cells mediate allergic inflammation within the context of the lung environment. Ultimately, we aim to highlight important future research questions that will lead to novel therapeutic strategies for fungal allergic diseases.
Collapse
Affiliation(s)
- Julio Furlong-Silva
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Peter Charles Cook
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
- * E-mail:
| |
Collapse
|
12
|
Wilson KR, Gressier E, McConville MJ, Bedoui S. Microbial Metabolites in the Maturation and Activation of Dendritic Cells and Their Relevance for Respiratory Immunity. Front Immunol 2022; 13:897462. [PMID: 35880171 PMCID: PMC9307905 DOI: 10.3389/fimmu.2022.897462] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/10/2022] [Indexed: 12/12/2022] Open
Abstract
The respiratory tract is a gateway for viruses and bacteria from the external environment to invade the human body. Critical to the protection against these invaders are dendritic cells (DCs) - a group of highly specialized myeloid cells that monitors the lung microenvironment and relays contextual and antigenic information to T cells. Following the recognition of danger signals and/or pathogen molecular associated patterns in the lungs, DCs undergo activation. This process arms DCs with the unique ability to induce the proliferation and differentiation of T cells responding to matching antigen in complex with MHC molecules. Depending on how DCs interact with T cells, the ensuing T cell response can be tolerogenic or immunogenic and as such, the susceptibility and severity of respiratory infections is influenced by the signals DCs receive, integrate, and then convey to T cells. It is becoming increasingly clear that these facets of DC biology are heavily influenced by the cellular components and metabolites produced by the lung and gut microbiota. In this review, we discuss the roles of different DC subsets in respiratory infections and outline how microbial metabolites impact the development, propensity for activation and subsequent activation of DCs. In particular, we highlight these concepts in the context of respiratory immunity.
Collapse
Affiliation(s)
- Kayla R. Wilson
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
- *Correspondence: Kayla R. Wilson,
| | - Elise Gressier
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Malcolm J. McConville
- Department of Biochemistry and Pharmacology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Melbourne, VIC, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
13
|
Klimova N, Holubova J, Streparola G, Tomala J, Brazdilova L, Stanek O, Bumba L, Sebo P. Pertussis toxin suppresses dendritic cell-mediated delivery of B. pertussis into lung-draining lymph nodes. PLoS Pathog 2022; 18:e1010577. [PMID: 35666769 PMCID: PMC9216613 DOI: 10.1371/journal.ppat.1010577] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/22/2022] [Accepted: 05/08/2022] [Indexed: 11/18/2022] Open
Abstract
The adenylate cyclase (ACT) and the pertussis (PT) toxins of Bordetella pertussis exert potent immunomodulatory activities that synergize to suppress host defense in the course of whooping cough pathogenesis. We compared the mouse lung infection capacities of B. pertussis (Bp) mutants (Bp AC− or Bp PT–) producing enzymatically inactive toxoids and confirm that ACT action is required for maximal bacterial proliferation in the first days of infection, whereas PT action is crucial for persistence of B. pertussis in mouse lungs. Despite accelerated and near complete clearance from the lungs by day 14 of infection, the PT− bacteria accumulated within the lymphoid tissue of lung-draining mediastinal lymph nodes (mLNs). In contrast, the wild type or AC− bacteria colonized the lungs but did not enter into mLNs. Lung infection by the PT− mutant triggered an early arrival of migratory conventional dendritic cells with associated bacteria into mLNs, where the PT− bacteria entered the T cell-rich paracortex of mLNs by day 5 and proliferated in clusters within the B-cell zone (cortex) of mLNs by day 14, being eventually phagocytosed by infiltrating neutrophils. Finally, only infection by the PT− bacteria triggered an early production of anti-B. pertussis serum IgG antibodies already within 14 days of infection. These results reveal that action of the pertussis toxin blocks DC-mediated delivery of B. pertussis bacteria into mLNs and prevents bacterial colonization of mLNs, thus hampering early adaptive immune response to B. pertussis infection. Of the three classical Bordetella species causing respiratory infections in mammals, only the human-specialized whooping cough agent B. pertussis produces the pertussis toxin (PT) as its major virulence factor. Human pertussis is an acute respiratory illness and the pleiotropic activities of pertussis toxin account for the characteristic systemic manifestations of the disease, such as hyperleukocytosis, histamine sensitization, hyperinsulinemia, or inflammatory lung pathology. We found that PT activity inhibits the migration of infected dendritic cells from the lungs into the draining mediastinal lymph nodes (mLNs). This prevents mLN infection by bacteria evading from migratory cells and delivery of bacterial antigens into mLNs. As a result, the induction of adaptive serum antibody responses to infection is delayed. We thus propose that PT action serves to create a time window for proliferation of B. pertussis on airway mucosa to facilitate transmission of the pathogen among humans.
Collapse
Affiliation(s)
- Nela Klimova
- Institute of Microbiology of the Czech Academy of Sciences,Prague, Czech Republic
- Faculty of Sciences, Charles University, Prague, Czech Republic
| | - Jana Holubova
- Institute of Microbiology of the Czech Academy of Sciences,Prague, Czech Republic
| | - Gaia Streparola
- Institute of Microbiology of the Czech Academy of Sciences,Prague, Czech Republic
- Czech Centre for Phenogenomics BIOCEV, Vestec, Czech Republic
| | - Jakub Tomala
- Institute of Microbiology of the Czech Academy of Sciences,Prague, Czech Republic
- Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Ludmila Brazdilova
- Institute of Microbiology of the Czech Academy of Sciences,Prague, Czech Republic
- Faculty of Sciences, Charles University, Prague, Czech Republic
| | - Ondrej Stanek
- Institute of Microbiology of the Czech Academy of Sciences,Prague, Czech Republic
| | - Ladislav Bumba
- Institute of Microbiology of the Czech Academy of Sciences,Prague, Czech Republic
- * E-mail: (LB); (PS)
| | - Peter Sebo
- Institute of Microbiology of the Czech Academy of Sciences,Prague, Czech Republic
- * E-mail: (LB); (PS)
| |
Collapse
|
14
|
Li R, Hu X, Chen H, Zhao Y, Gao X, Yuan Y, Guo H, Huang H, Zou X, Qi H, Liu H, Shang Y. Role of Cholinergic Anti-Inflammatory Pathway in Protecting Sepsis-Induced Acute Lung Injury through Regulation of the Conventional Dendritic Cells. Mediators Inflamm 2022; 2022:1474891. [PMID: 35125962 PMCID: PMC8813293 DOI: 10.1155/2022/1474891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/03/2022] [Accepted: 01/06/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The cholinergic anti-inflammatory pathway connects the immune response system and the nervous system via the vagus nerve. The key regulatory receptor is the α7-subtype of the nicotinic acetylcholine receptor (α7nAChR). Cholinergic anti-inflammatory pathway has been proved to be effective in suppressing the inflammation responses in acute lung injury (ALI). Dendritic cells (DCs), the important antigen-presenting cells, also express the α7nAChR. Past studies have indicated that reducing the quantity of mature conventional DCs and inhibiting the maturation of pulmonary DCs may prove effective for the treatment of ALI. However, the effects of cholinergic anti-inflammatory pathway on maturation, function, and quantity of DCs and conventional DCs in ALI remain unclear. OBJECTIVE It was hypothesized that cholinergic anti-inflammatory pathway may inhibit the inflammatory response of ALI by regulating maturation, phenotype, and quantity of DCs and conventional DCs. METHODS GTS-21 (GTS-21 dihydrochloride), an α7nAchR agonist, was prophylactically administered in sepsis-induced ALI mouse model and LPS-primed bone marrow-derived dendritic cells. The effects of GTS-21 were observed with respect to maturation, phenotype, and quantity of DCs, conventional DCs, and conventional DCs2 (type 2 conventional DCs) and the release of DC-related proinflammatory cytokines in vivo and in vitro. RESULTS The results of the present study revealed that GTS-21 treatment decreased the maturation of DCs and the production of DC-related proinflammatory cytokines in vitro and in sepsis-induced ALI mouse model; it reduced the quantity of CD11c+MHCII+ conventional DCs and CD11c+CD11b+ conventional DCs2 in vivo experiment. CONCLUSIONS Cholinergic anti-inflammatory pathway contributes to the reduction in the inflammatory response in ALI by regulating maturation, phenotype, and quantity of DCs, conventional DCs, and conventional DCs2.
Collapse
Affiliation(s)
- Ruiting Li
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xuemei Hu
- Department of Nephrology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province 442000, China
| | - Huibin Chen
- Department of Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province 442000, China
| | - Yue Zhao
- Department of Critical Care Medicine, Jin Yin-tan Hospital, Wuhan, Hubei 430048, China
| | - Xuehui Gao
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Yin Yuan
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Huiling Guo
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Haiyan Huang
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xiaojing Zou
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Hong Qi
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Hong Liu
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - You Shang
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| |
Collapse
|
15
|
Sun X, Perl AK, Li R, Bell SM, Sajti E, Kalinichenko VV, Kalin TV, Misra RS, Deshmukh H, Clair G, Kyle J, Crotty Alexander LE, Masso-Silva JA, Kitzmiller JA, Wikenheiser-Brokamp KA, Deutsch G, Guo M, Du Y, Morley MP, Valdez MJ, Yu HV, Jin K, Bardes EE, Zepp JA, Neithamer T, Basil MC, Zacharias WJ, Verheyden J, Young R, Bandyopadhyay G, Lin S, Ansong C, Adkins J, Salomonis N, Aronow BJ, Xu Y, Pryhuber G, Whitsett J, Morrisey EE. A census of the lung: CellCards from LungMAP. Dev Cell 2022; 57:112-145.e2. [PMID: 34936882 PMCID: PMC9202574 DOI: 10.1016/j.devcel.2021.11.007] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/19/2021] [Accepted: 11/05/2021] [Indexed: 01/07/2023]
Abstract
The human lung plays vital roles in respiration, host defense, and basic physiology. Recent technological advancements such as single-cell RNA sequencing and genetic lineage tracing have revealed novel cell types and enriched functional properties of existing cell types in lung. The time has come to take a new census. Initiated by members of the NHLBI-funded LungMAP Consortium and aided by experts in the lung biology community, we synthesized current data into a comprehensive and practical cellular census of the lung. Identities of cell types in the normal lung are captured in individual cell cards with delineation of function, markers, developmental lineages, heterogeneity, regenerative potential, disease links, and key experimental tools. This publication will serve as the starting point of a live, up-to-date guide for lung research at https://www.lungmap.net/cell-cards/. We hope that Lung CellCards will promote the community-wide effort to establish, maintain, and restore respiratory health.
Collapse
Affiliation(s)
- Xin Sun
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | - Anne-Karina Perl
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Rongbo Li
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Sheila M Bell
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Eniko Sajti
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Vladimir V Kalinichenko
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA; Center for Lung Regenerative Medicine, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Tanya V Kalin
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Ravi S Misra
- Department of Pediatrics Division of Neonatology, The University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Hitesh Deshmukh
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Geremy Clair
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Jennifer Kyle
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Laura E Crotty Alexander
- Deparment of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jorge A Masso-Silva
- Deparment of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Joseph A Kitzmiller
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Kathryn A Wikenheiser-Brokamp
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pathology & Laboratory Medicine, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Gail Deutsch
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA; Department of Laboratories, Seattle Children's Hospital, OC.8.720, 4800 Sand Point Way Northeast, Seattle, WA 98105, USA
| | - Minzhe Guo
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Yina Du
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Michael P Morley
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J Valdez
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Haoze V Yu
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Kang Jin
- Departments of Biomedical Informatics, Developmental Biology, and Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Eric E Bardes
- Departments of Biomedical Informatics, Developmental Biology, and Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jarod A Zepp
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Terren Neithamer
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maria C Basil
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William J Zacharias
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Internal Medicine, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Jamie Verheyden
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Randee Young
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Gautam Bandyopadhyay
- Department of Pediatrics Division of Neonatology, The University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Sara Lin
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Charles Ansong
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Joshua Adkins
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Nathan Salomonis
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA; Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Bruce J Aronow
- Departments of Biomedical Informatics, Developmental Biology, and Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yan Xu
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Gloria Pryhuber
- Department of Pediatrics Division of Neonatology, The University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jeff Whitsett
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Edward E Morrisey
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
16
|
Kozlovski S, Regev O, Sapoznikov A, Kizner M, Achdout H, Petrovich-Kopitman E, Elkahal J, Addadi Y, Silva Castanheira FVE, Feigelson SW, Kubes P, Erez N, Garbi N, Alon R. ICAMs are dispensable for influenza clearance and anti-viral humoral and cellular immunity. Front Immunol 2022; 13:1041552. [PMID: 36895258 PMCID: PMC9988921 DOI: 10.3389/fimmu.2022.1041552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 12/12/2022] [Indexed: 02/25/2023] Open
Abstract
αLβ2 (LFA-1) mediated interactions with ICAM-1 and ICAM-2 predominate leukocyte-vascular interactions, but their functions in extravascular cell-cell communications is still debated. The roles of these two ligands in leukocyte trafficking, lymphocyte differentiation, and immunity to influenza infections were dissected in the present study. Surprisingly, double ICAM-1 and ICAM-2 knock out mice (herein ICAM-1/2-/- mice) infected with a lab adapted H1N1 influenza A virus fully recovered from infection, elicited potent humoral immunity, and generated normal long lasting anti-viral CD8+ T cell memory. Furthermore, lung capillary ICAMs were dispensable for both NK and neutrophil entry to virus infected lungs. Mediastinal lymph nodes (MedLNs) of ICAM-1/2-/- mice poorly recruited naïve T cells and B lymphocytes but elicited normal humoral immunity critical for viral clearance and effective CD8+ differentiation into IFN-γ producing T cells. Furthermore, whereas reduced numbers of virus specific effector CD8+ T cells accumulated inside infected ICAM-1/2-/- lungs, normal virus-specific TRM CD8+ cells were generated inside these lungs and fully protected ICAM-1/2-/- mice from secondary heterosubtypic infections. B lymphocyte entry to the MedLNs and differentiation into extrafollicular plasmablasts, producing high affinity anti-influenza IgG2a antibodies, were also ICAM-1 and ICAM-2 independent. A potent antiviral humoral response was associated with accumulation of hyper-stimulated cDC2s in ICAM null MedLNs and higher numbers of virus-specific T follicular helper (Tfh) cells generated following lung infection. Mice selectively depleted of cDC ICAM-1 expression supported, however, normal CTL and Tfh differentiation following influenza infection, ruling out essential co-stimulatory functions of DC ICAM-1 in CD8+ and CD4+ T cell differentiation. Collectively our findings suggest that lung ICAMs are dispensable for innate leukocyte trafficking to influenza infected lungs, for the generation of peri-epithelial TRM CD8+ cells, and long term anti-viral cellular immunity. In lung draining LNs, although ICAMs promote lymphocyte homing, these key integrin ligands are not required for influenza-specific humoral immunity or generation of IFN-γ effector CD8+ T cells. In conclusion, our findings suggest unexpected compensatory mechanisms that orchestrate protective anti-influenza immunity in the absence of vascular and extravascular ICAMs.
Collapse
Affiliation(s)
- Stav Kozlovski
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ofer Regev
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Anita Sapoznikov
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Marina Kizner
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hagit Achdout
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | | | - Jacob Elkahal
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yoseph Addadi
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | | | - Sara W Feigelson
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Paul Kubes
- Department of Pharmacology and Physiology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Noam Erez
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Natalio Garbi
- Department of Cellular Immunology, Institute of Experimental Immunology Medical Faculty, University of Bonn, Bonn, Germany
| | - Ronen Alon
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
17
|
Monophosphoryl Lipid A and Poly I:C Combination Adjuvant Promoted Ovalbumin-Specific Cell Mediated Immunity in Mice Model. BIOLOGY 2021; 10:biology10090908. [PMID: 34571785 PMCID: PMC8471534 DOI: 10.3390/biology10090908] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/10/2021] [Accepted: 09/10/2021] [Indexed: 01/22/2023]
Abstract
Simple Summary Many research groups have investigated and developed new adjuvant candidates to promote vaccine efficacy, but only few of them were licensed. A combination of toll-like receptor (TLR) agonists can be a promising vaccine adjuvant candidate by stimulating innate immune cells and inducing antigen-specific cell-mediated immunity (CMI). In this study, a monophosphoryl lipid A (MPL) and Poly I:C combination, in low doses with ovalbumin (OVA) protein, elicited strong OVA-specific antibody production and more effective memory T cell responses compared with OVA only, OVA+MPL, OVA+Poly I:C groups at the site of immunization, as well as innate immune cell recruitment and activation. This study suggests MPL+Poly I:C as a potential CMI-inducing vaccine adjuvant candidate. Abstract Induction of antigen-specific cell-mediated immunity (CMI), as well as humoral immunity, is critical for successful vaccination against various type of pathogens. Toll-like receptor (TLR) agonists have been developed as adjuvants to promote vaccine efficacy and induce appropriate immune responses. Monophosphoryl lipid A (MPL); a TLR4 agonist, and Poly I:C; a TLR3 agonist, are known as a strong immuno-stimulator which induce Th1 response. Many studies proved and compared the efficacy of each adjuvant, but no study has investigated the combination of them. Using ovalbumin protein antigen, MPL+Poly I:C combination induced more effective antigen-specific CMI response than single adjuvants. Production of inflammatory cytokines, recruitment of innate immune cells and antigen-specific CD4/CD8 memory T cell at the immunized site had been significantly enhanced by MPL+Poly I:C combination. Moreover, MPL+Poly I:C combination enhanced ovalbumin-specific serum IgG, IgG1, and IgG2c production and proliferative function of CD4 and CD8 T cells after in vitro ovalbumin peptide stimulation. Taken together, these data suggest that the combination of MPL and Poly I:C has a potency as a CMI-inducing vaccine adjuvant with synergistically increased effects.
Collapse
|
18
|
Balhara J, Koussih L, Mohammed A, Shan L, Lamkhioued B, Gounni AS. PTX3 Deficiency Promotes Enhanced Accumulation and Function of CD11c +CD11b + DCs in a Murine Model of Allergic Inflammation. Front Immunol 2021; 12:641311. [PMID: 34305885 PMCID: PMC8299994 DOI: 10.3389/fimmu.2021.641311] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/21/2021] [Indexed: 11/17/2022] Open
Abstract
PTX3 is a unique member of the long pentraxins family and plays an indispensable role in regulating the immune system. We previously showed that PTX3 deletion aggravates allergic inflammation via a Th17 -dominant phenotype and enhanced CD4 T cell survival using a murine model of ovalbumin (OVA) induced allergic inflammation. In this study, we identified that upon OVA exposure, increased infiltration of CD11c+CD11b+ dendritic cells (DCs) was observed in the lungs of PTX3-/- mice compared to wild type littermate. Further analysis showed that a short-term OVA exposure led to an increased number of bone marrow common myeloid progenitors (CMP) population concomitantly with increased Ly6Chigh CCR2high monocytes and CD11c+CD11b+ DCs in the lungs. Also, pulmonary CD11c+CD11b+ DCs from OVA-exposed PTX3-/- mice exhibited enhanced expression of maturation markers, chemokines receptors CCR2, and increased OVA uptake and processing compared to wild type controls. Taken together, our data suggest that PTX3 deficiency heightened lung CD11c+CD11b+DC numbers and function, hence exacerbating airway inflammatory response.
Collapse
Affiliation(s)
- Jyoti Balhara
- Department of Immunology, Max-Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Latifa Koussih
- Department of Immunology, Max-Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.,Department des Sciences Experimentales, Université de Saint-Boniface, Winnipeg, MB, Canada
| | - Ashfaque Mohammed
- Department of Immunology, Max-Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Lianyu Shan
- Department of Immunology, Max-Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Bouchaib Lamkhioued
- Laboratoire d'Immunologie et de Biotechnologies, EA7509-IRMAIC, Pôle-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Abdelilah S Gounni
- Department of Immunology, Max-Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
19
|
Pinto A, Rega A, Crother TR, Sorrentino R. Plasmacytoid dendritic cells and their therapeutic activity in cancer. Oncoimmunology 2021; 1:726-734. [PMID: 22934264 PMCID: PMC3429576 DOI: 10.4161/onci.20171] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In the last decade several studies provided evidence that plasmacytoid dendritic cells (pDCs) infiltrate human neoplasms with poor prognosis. However, the role of tumor-associated pDCs remains controversial. Various studies indicate that pDCs play an immuno-suppressive role and facilitate tumor progression in both animal models and humans. In contrast, others found that the presence of activated tumor-associated pDCs results in tumor regression in mice. Given these findings, understanding pDC function in tumor biology is an important necessity and may pave the way for novel therapeutic strategies to fight malignancies.
Collapse
Affiliation(s)
- Aldo Pinto
- Pharmaceutical and Biomedical Sciences Department (FARMABIOMED); University of Salerno; Fisciano, Italy
| | | | | | | |
Collapse
|
20
|
Morianos I, Semitekolou M. Dendritic Cells: Critical Regulators of Allergic Asthma. Int J Mol Sci 2020; 21:ijms21217930. [PMID: 33114551 PMCID: PMC7663753 DOI: 10.3390/ijms21217930] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 10/15/2020] [Accepted: 10/25/2020] [Indexed: 12/17/2022] Open
Abstract
Allergic asthma is a chronic inflammatory disease of the airways characterized by airway hyperresponsiveness (AHR), chronic airway inflammation, and excessive T helper (Th) type 2 immune responses against harmless airborne allergens. Dendritic cells (DCs) represent the most potent antigen-presenting cells of the immune system that act as a bridge between innate and adaptive immunity. Pertinent to allergic asthma, distinct DC subsets are known to play a central role in initiating and maintaining allergen driven Th2 immune responses in the airways. Nevertheless, seminal studies have demonstrated that DCs can also restrain excessive asthmatic responses and thus contribute to the resolution of allergic airway inflammation and the maintenance of pulmonary tolerance. Notably, the transfer of tolerogenic DCs in vivo suppresses Th2 allergic responses and protects or even reverses established allergic airway inflammation. Thus, the identification of novel DC subsets that possess immunoregulatory properties and can efficiently control aberrant asthmatic responses is critical for the re-establishment of tolerance and the amelioration of the asthmatic disease phenotype.
Collapse
|
21
|
Troy A, Esparza-Gonzalez SC, Bartek A, Creissen E, Izzo L, Izzo AA. Pulmonary mucosal immunity mediated through CpG provides adequate protection against pulmonary Mycobacterium tuberculosis infection in the mouse model. A role for type I interferon. Tuberculosis (Edinb) 2020; 123:101949. [PMID: 32741537 DOI: 10.1016/j.tube.2020.101949] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/08/2020] [Accepted: 05/10/2020] [Indexed: 12/28/2022]
Abstract
Toll-Like Receptor (TLR) 9 stimulation is required for induction of potent immune responses against pathogen invasion. The use of unmethylated CpG as adjuvants in vaccines provides an excellent means of stimulating adaptive immunity. Our data demonstrate that CpG-C provided prolonged immune responses in the mouse model of tuberculosis when formulated with liposomes and the Mycobacterium tuberculosis antigen ESAT-6. A reduction in the mycobacterial burden was best achieved when administered as an intranasal vaccine and was dependent on type I interferon (IFN). There was a significant difference between CpG-C inoculated wild type and IFN-αR1-/- mice, indicating that type I IFN plays a role in the immune response following CpG-C inoculation. Further analysis showed that early NK cell presence was not an absolute requirement, although elevated IFN-γ levels were detected in the lungs of mice within 48 h. The reduction in mycobacterial burden was MyD88-independent as CpG-C inoculated MyD88-/- mice showed comparable mycobacterial burdens to wild type mice with no detriment due to the lack of MyD88. Together our data show that pulmonary stimulation of TLR9 bearing antigen presenting cells resulted in the induction of protective immunity against M. tuberculosis infection that was dependent on type I IFN signaling.
Collapse
Affiliation(s)
- Amber Troy
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| | - Sandra C Esparza-Gonzalez
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| | - Alicia Bartek
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| | - Elizabeth Creissen
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| | - Linda Izzo
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| | - Angelo A Izzo
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA.
| |
Collapse
|
22
|
Pulmonary CD103+ dendritic cells: key regulators of immunity against infection. Cell Mol Immunol 2020; 17:670-671. [PMID: 32203184 DOI: 10.1038/s41423-020-0397-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/19/2022] Open
|
23
|
Barjesteh N, O'Dowd K, Vahedi SM. Antiviral responses against chicken respiratory infections: Focus on avian influenza virus and infectious bronchitis virus. Cytokine 2020; 127:154961. [PMID: 31901597 PMCID: PMC7129915 DOI: 10.1016/j.cyto.2019.154961] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 12/13/2022]
Abstract
Some of the respiratory viral infections in chickens pose a significant threat to the poultry industry and public health. In response to viral infections, host innate responses provide the first line of defense against viruses, which often act even before the establishment of the infection. Host cells sense the presence of viral components through germinal encoded pattern recognition receptors (PRRs). The engagement of PRRs with pathogen-associated molecular patterns leads to the induction of pro-inflammatory and interferon productions. Induced antiviral responses play a critical role in the outcome of the infections. In order to improve current strategies for control of viral infections or to advance new strategies aimed against viral infections, a deep understanding of host-virus interaction and induction of antiviral responses is required. In this review, we summarized recent progress in understanding innate antiviral responses in chickens with a focus on the avian influenza virus and infectious bronchitis virus.
Collapse
Affiliation(s)
- Neda Barjesteh
- Research Group on Infectious Diseases in Production Animals (GREMIP), and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada.
| | - Kelsey O'Dowd
- Research Group on Infectious Diseases in Production Animals (GREMIP), and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Seyed Milad Vahedi
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
24
|
Wu HM, Zhao CC, Xie QM, Xu J, Fei GH. TLR2-Melatonin Feedback Loop Regulates the Activation of NLRP3 Inflammasome in Murine Allergic Airway Inflammation. Front Immunol 2020; 11:172. [PMID: 32117301 PMCID: PMC7025476 DOI: 10.3389/fimmu.2020.00172] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 01/22/2020] [Indexed: 12/13/2022] Open
Abstract
Toll-like receptor 2 (TLR2) is suggested to initiate the activation of NLRP3 inflammasome, and considered to be involved in asthma. The findings that melatonin modulates TLRs-mediated immune responses, together with the suppressing effect of TLRs on endogenous melatonin synthesis, support the possibility that a feedback loop exists between TLRs system and endogenous melatonin synthesis. To determine whether TLR2-melatonin feedback loop exists in allergic airway disease and regulates NLRP3 inflammasome activity, wild-type (WT) and TLR2−/− mice were challenged with OVA to establish allergic airway disease model. Following OVA challenge, WT mice exhibited increased-expression of TLR2, activation of NLRP3 inflammasome and marked airway inflammation, which were all effectively inhibited in the TLR2−/− mice, indicating that TLR2-NLRP3 mediated airway inflammation. Meanwhile, melatonin biosynthesis was reduced in OVA-challenged WT mice, while such reduction was notably rescued by TLR2 deficiency, suggesting that TLR2-NLRP3-mediated allergic airway inflammation was associated with decreased endogenous melatonin biosynthesis. Furthermore, addition of melatonin to OVA-challenged WT mice pronouncedly ameliorated airway inflammation, decreased TLR2 expression and NLRP3 inflammasome activation, further implying that melatonin in turn inhibited airway inflammation via suppressing TLR2-NLRP3 signal. Most interestingly, although melatonin receptor antagonist luzindole significantly reduced the protein expressions of ASMT, AANAT and subsequent level of melatonin in OVA-challenged TLR2−/− mice, it exhibited null effect on leukocytes infiltration, Th2-cytokines production and NLRP3 activity. These results indicate that a TLR2-melatonin feedback loop regulates NLRP3 inflammasome activity in allergic airway inflammation, and melatonin may be a promising therapeutic medicine for airway inflammatory diseases such as asthma.
Collapse
Affiliation(s)
- Hui-Mei Wu
- Department of Geriatric Respiratory and Critical Care, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Key Laboratory of Geriatric Molecular Medicine, Anhui Medical University, Hefei, China
| | - Cui-Cui Zhao
- Department of Geriatric Respiratory and Critical Care, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Key Laboratory of Geriatric Molecular Medicine, Anhui Medical University, Hefei, China
| | - Qiu-Meng Xie
- Department of Geriatric Respiratory and Critical Care, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Key Laboratory of Geriatric Molecular Medicine, Anhui Medical University, Hefei, China
| | - Juan Xu
- Department of Geriatric Respiratory and Critical Care, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Key Laboratory of Geriatric Molecular Medicine, Anhui Medical University, Hefei, China
| | - Guang-He Fei
- Department of Respiratory and Critical Care, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
25
|
Liang W, Chen K, Gong W, Yoshimura T, Le Y, Wang Y, Wang JM. The Contribution of Chemoattractant GPCRs, Formylpeptide Receptors, to Inflammation and Cancer. Front Endocrinol (Lausanne) 2020; 11:17. [PMID: 32038501 PMCID: PMC6993212 DOI: 10.3389/fendo.2020.00017] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/09/2020] [Indexed: 12/14/2022] Open
Abstract
A hallmark of inflammatory responses is leukocyte mobilization, which is mediated by pathogen and host released chemotactic factors that activate Gi-protein-coupled seven-transmembrane receptors (GPCRs) on host cell surface. Formylpeptide receptors (FPRs, Fprs in mice) are members of the chemoattractant GPCR family, shown to be critical in myeloid cell trafficking during infection, inflammation, immune responses, and cancer progression. Accumulating evidence demonstrates that both human FPRs and murine Fprs are involved in a number of patho-physiological processes because of their expression on a wide variety of cell types in addition to myeloid cells. The unique capacity of FPRs (Fprs) to interact with numerous structurally unrelated chemotactic ligands enables these receptors to participate in orchestrated disease initiation, progression, and resolution. One murine Fpr member, Fpr2, and its endogenous agonist peptide, Cathelicidin-related antimicrobial peptide (CRAMP), have been demonstrated as key mediators of colon mucosal homeostasis and protection from inflammation and associated tumorigenesis. Recent availability of genetically engineered mouse models greatly expanded the understanding of the role of FPRs (Fprs) in pathophysiology that places these molecules in the list of potential targets for therapeutic intervention of diseases.
Collapse
Affiliation(s)
- Weiwei Liang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Keqiang Chen
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Teizo Yoshimura
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yingying Le
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Ying Wang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Ji Ming Wang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| |
Collapse
|
26
|
Palomino-Segura M, Perez L, Farsakoglu Y, Virgilio T, Latino I, D'Antuono R, Chatziandreou N, Pizzagalli DU, Wang G, García-Sastre A, Sallusto F, Carroll MC, Neyrolles O, Gonzalez SF. Protection against influenza infection requires early recognition by inflammatory dendritic cells through C-type lectin receptor SIGN-R1. Nat Microbiol 2019; 4:1930-1940. [PMID: 31358982 PMCID: PMC6817362 DOI: 10.1038/s41564-019-0506-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 06/06/2019] [Indexed: 12/23/2022]
Abstract
The early phase of influenza infection occurs in the upper respiratory tract and the trachea, but little is known about the initial events of virus recognition and control of viral dissemination by the immune system. Here, we report that inflammatory dendritic cells (IDCs) are recruited to the trachea shortly after influenza infection through type I interferon-mediated production of the chemokine CCL2. We further show that recruited IDCs express the C-type lectin receptor SIGN-R1, which mediates direct recognition of the virus by interacting with N-linked glycans present in glycoproteins of the virion envelope. Activation of IDCs via SIGN-R1 triggers the production of the chemokines CCL5, CXCL9 and CXCL10, which initiate the recruitment of protective natural killer (NK) cells in the infected trachea. In the absence of SIGN-R1, the recruitment and activation of NK cells is impaired, leading to uncontrolled viral proliferation. In sum, our results provide insight into the orchestration of the early cellular and molecular events involved in immune protection against influenza.
Collapse
Affiliation(s)
- Miguel Palomino-Segura
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Graduate School of Cellular and Molecular Sciences, Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Laurent Perez
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Yagmur Farsakoglu
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Graduate School of Cellular and Molecular Sciences, Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Tommaso Virgilio
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Graduate School of Cellular and Molecular Sciences, Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Irene Latino
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Rocco D'Antuono
- Light Microscopy STP, The Francis Crick Institute, London, UK
| | - Nikolaos Chatziandreou
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Diego U Pizzagalli
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Computational Science, Università della Svizzera italiana, Lugano, Switzerland
| | - Guojun Wang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogen Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogen Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute for Microbiology, ETH Zurich, Zurich, Switzerland
| | - Michael C Carroll
- Program in Cellular and Molecular Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse CNRS, UPS, Toulouse, France
| | - Santiago F Gonzalez
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland.
| |
Collapse
|
27
|
Beyeler S, Steiner S, Wotzkow C, Tschanz SA, Adhanom Sengal A, Wick P, Haenni B, Alves MP, von Garnier C, Blank F. Multi-walled carbon nanotubes activate and shift polarization of pulmonary macrophages and dendritic cells in an in vivo model of chronic obstructive lung disease. Nanotoxicology 2019; 14:77-96. [PMID: 31556347 DOI: 10.1080/17435390.2019.1663954] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
With substantial progress of nanotechnology, there is rising concern about possible adverse health effects related to inhalation of nanomaterials, such as multi-walled carbon nanotubes (MWCNT). In particular, individuals with chronic respiratory disorders, such as chronic obstructive pulmonary disease (COPD), may potentially be more susceptible to adverse health effects related to inhaled MWCNT. Hazard assessment of such inhaled nanomaterials therefore requires timely clarification. This was assessed in this study using a mouse model of COPD by exposing animals to 0.08 µg/cm2 of MWCNT administered by intratracheal instillation. Treatment with MWCNT induced an accumulation of alveolar macrophages (AMφ) in bronchoalveolar lavage fluid (BALF) in COPD mice that increased from 24 h to 7 d. In COPD mice, MWCNT induced a dynamic shift in macrophage polarization as measured by expression of CD38 and CD206, and increased AMφ and lung parenchyma macrophage (LPMΦ) activation with upregulation of co-stimulatory markers CD40 and CD80. Moreover, MWCNT treatment increased the frequencies of pulmonary dendritic cells (DC), leading to an expansion of the CD11b+CD103- DC subset. Although MWCNT did not trigger lung functional or structural changes, they induced an increased expression of the muc5AC transcript in mice with COPD. Our data provide initial evidence that inhaled MWCNT affect the pulmonary mucosal immune system by altering the numbers, phenotype, and activation status of antigen-presenting cell populations. Extrapolating these in vivo mouse findings to human pulmonary MWCNT exposure, caution is warranted in limiting exposure when handling inhalable nanofibers.
Collapse
Affiliation(s)
- Seraina Beyeler
- Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Selina Steiner
- Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Carlos Wotzkow
- Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | | | - Amanuel Adhanom Sengal
- Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland
| | - Peter Wick
- Laboratory for Particles-Biology Interactions, Empa Materials Science and Technology, St. Gallen, Switzerland
| | - Beat Haenni
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Marco P Alves
- Institute of Virology and Immunology, Bern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Christophe von Garnier
- Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland
| | - Fabian Blank
- Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland
| |
Collapse
|
28
|
Arora S, Ahmad S, Irshad R, Goyal Y, Rafat S, Siddiqui N, Dev K, Husain M, Ali S, Mohan A, Syed MA. TLRs in pulmonary diseases. Life Sci 2019; 233:116671. [PMID: 31336122 PMCID: PMC7094289 DOI: 10.1016/j.lfs.2019.116671] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/16/2019] [Accepted: 07/19/2019] [Indexed: 12/21/2022]
Abstract
Toll-like receptors (TLRs) comprise a clan of proteins involved in identification and triggering a suitable response against pathogenic attacks. As lung is steadily exposed to multiple infectious agents, antigens and host-derived danger signals, the inhabiting stromal and myeloid cells of the lung express an aggregate of TLRs which perceive the endogenously derived damage-associated molecular patterns (DAMPs) along with pathogen associated molecular patterns (PAMPs) and trigger the TLR-associated signalling events involved in host defence. Thus, they form an imperative component of host defence activation in case of microbial infections as well as non-infectious pulmonary disorders such as interstitial lung disease, acute lung injury and airways disease, such as COPD and asthma. They also play an equally important role in lung cancer. Targeting the TLR signalling network would pave ways to the design of more reliable and effective vaccines against infectious agents and control deadly infections, desensitize allergens and reduce inflammation. Moreover, TLR agonists may act as adjuvants by increasing the efficiency of cancer vaccines, thereby contributing their role in treatment of lung cancer too. Overall, TLRs present a compelling and expeditiously bolstered area of research and addressing their signalling events would be of significant use in pulmonary diseases.
Collapse
Affiliation(s)
- Shweta Arora
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Shaniya Ahmad
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Rasha Irshad
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Yamini Goyal
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Sahar Rafat
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Neha Siddiqui
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Kapil Dev
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Mohammad Husain
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Shakir Ali
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India.
| | - Anant Mohan
- Department of Pulmonary Medicine, AIIMS, New Delhi, India.
| | - Mansoor Ali Syed
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| |
Collapse
|
29
|
Obieglo K, Schuijs MJ, Ozir-Fazalalikhan A, Otto F, van Wijck Y, Boon L, Lambrecht BN, Taube C, Smits HH. Isolated Schistosoma mansoni eggs prevent allergic airway inflammation. Parasite Immunol 2018; 40:e12579. [PMID: 30107039 PMCID: PMC6175163 DOI: 10.1111/pim.12579] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 07/30/2018] [Indexed: 12/26/2022]
Abstract
Chronic helminth infection with Schistosoma (S.) mansoni protects against allergic airway inflammation (AAI) in mice and is associated with reduced Th2 responses to inhaled allergens in humans, despite the presence of schistosome‐specific Th2 immunity. Schistosome eggs strongly induce type 2 immunity and allow to study the dynamics of Th2 versus regulatory responses in the absence of worms. Treatment with isolated S. mansoni eggs by i.p. injection prior to induction of AAI to ovalbumin (OVA)/alum led to significantly reduced AAI as assessed by less BAL and lung eosinophilia, less cellular influx into lung tissue, less OVA‐specific Th2 cytokines in lungs and lung‐draining mediastinal lymph nodes and less circulating allergen‐specific IgG1 and IgE antibodies. While OVA‐specific Th2 responses were inhibited, treatment induced a strong systemic Th2 response to the eggs. The protective effect of S. mansoni eggs was unaltered in μMT mice lacking mature (B2) B cells and unaffected by Treg cell depletion using anti‐CD25 blocking antibodies during egg treatment and allergic sensitization. Notably, prophylactic egg treatment resulted in a reduced influx of pro‐inflammatory, monocyte‐derived dendritic cells into lung tissue of allergic mice following challenge. Altogether, S. mansoni eggs can protect against the development of AAI, despite strong egg‐specific Th2 responses.
Collapse
Affiliation(s)
- Katja Obieglo
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Martijn J Schuijs
- Immunoregulation and Mucosal Immunology, VIB Inflammation Research Center, Ghent, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium
| | | | - Frank Otto
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Yolanda van Wijck
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Bart N Lambrecht
- Immunoregulation and Mucosal Immunology, VIB Inflammation Research Center, Ghent, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Christian Taube
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
30
|
Batf3-Dependent Dendritic Cells Promote Optimal CD8 T Cell Responses against Respiratory Poxvirus Infection. J Virol 2018; 92:JVI.00495-18. [PMID: 29875235 DOI: 10.1128/jvi.00495-18] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/23/2018] [Indexed: 02/06/2023] Open
Abstract
Respiratory infection with vaccinia virus (VacV) elicits robust CD8+ T cell responses that play an important role in host resistance. In the lung, VacV encounters multiple tissue-resident antigen-presenting cell (APC) populations, but which cell plays a dominant role in priming of virus-specific CD8+ effector T cell responses remains poorly defined. We used Batf3-/- mice to investigate the impact of CD103+ and CD8α+ dendritic cell (DC) deficiency on anti-VacV CD8+ T cell responses. We found that Batf3-/- mice were more susceptible to VacV infection, exhibiting profound weight loss, which correlated with impaired accumulation of gamma interferon (IFN-γ)-producing CD8+ T cells in the lungs. This was largely due to defective priming since early in the response, antigen-specific CD8+ T cells in the draining lymph nodes of Batf3-/- mice expressed significantly reduced levels of Ki67, CD25, and T-bet. These results underscore a specific role for Batf3-dependent DCs in regulating priming and expansion of effector CD8+ T cells necessary for host resistance against acute respiratory VacV infection.IMPORTANCE During respiratory infection with vaccinia virus (VacV), a member of Poxviridae family, CD8+ T cells play important role in resolving the primary infection. Effector CD8+ T cells clear the virus by accumulating in the infected lungs in large numbers and secreting molecules such as IFN-γ that kill virally infected cells. However, precise cell types that regulate the generation of effector CD8+ T cells in the lungs are not well defined. Dendritic cells (DCs) are a heterogeneous population of immune cells that are recognized as key initiators and regulators of T-cell-mediated immunity. In this study, we reveal that a specific subset of DCs that are dependent on the transcription factor Batf3 for their development regulate the magnitude of CD8+ T cell effector responses in the lungs, thereby providing protection during pulmonary VacV infection.
Collapse
|
31
|
CD4 T Cell Epitope Specificity and Cytokine Potential Are Preserved as Cells Transition from the Lung Vasculature to Lung Tissue following Influenza Virus Infection. J Virol 2018; 92:JVI.00377-18. [PMID: 29669836 DOI: 10.1128/jvi.00377-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 04/13/2018] [Indexed: 01/21/2023] Open
Abstract
Pulmonary CD4 T cells are critical in respiratory virus control, both by delivering direct effector function and through coordinating responses of other immune cells. Recent studies have shown that following influenza virus infection, virus-specific CD4 T cells are partitioned between pulmonary vasculature and lung tissue. However, very little is known about the peptide specificity or functional differences of CD4 T cells within these two compartments. Using a mouse model of influenza virus infection in conjunction with intravascular labeling in vivo, the cell surface phenotype, epitope specificity, and functional potential of the endogenous polyclonal CD4 T cell response was examined by tracking nine independent CD4 T cell epitope specificities. These studies revealed that tissue-localized CD4 cells were globally distinct from vascular cells in expression of markers associated with transendothelial migration, residency, and micropositioning. Despite these differences, there was little evidence for remodeling of the viral epitope specificity or cytokine potential as cells transition from vasculature to the highly inflamed lung tissue. Our studies also distinguished cells in the pulmonary vasculature from peripheral circulating CD4 T cells, providing support for the concept that the pulmonary vasculature does not simply reflect circulating cells that are trapped within the narrow confines of capillary vessels but rather is enriched in transitional cells primed in the draining lymph node that have specialized potential to enter the lung tissue.IMPORTANCE CD4 T cells convey a multitude of functions in immunity to influenza, including those delivered in the lymph node and others conveyed by CD4 T cells that leave the lymph node, enter the blood, and extravasate into the lung tissue. Here, we show that the transition of recently primed CD4 cells detected in the lung vasculature undergo profound changes in expression of markers associated with tissue localization as they establish residence in the lung. However, this transition does not edit CD4 T cell epitope specificity or the cytokine potential of the CD4 T cells. Thus, CD4 T cells that enter the infected lung can convey diverse functions and have a sufficiently broad viral antigen specificity to detect the complex array of infected cells within the infected tissue, offering the potential for more effective protective function.
Collapse
|
32
|
Ko EJ, Lee Y, Lee YT, Kim YJ, Kim KH, Kang SM. MPL and CpG combination adjuvants promote homologous and heterosubtypic cross protection of inactivated split influenza virus vaccine. Antiviral Res 2018; 156:107-115. [PMID: 29885376 DOI: 10.1016/j.antiviral.2018.06.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 06/02/2018] [Accepted: 06/05/2018] [Indexed: 11/24/2022]
Abstract
Annual vaccination is not effective in conferring cross-protection against antigenically different influenza viruses. Therefore, it is of high priority to improve the cross protective efficacy of influenza vaccines. We investigated the adjuvant effects of monophosphoryl lipid A (MPL) and oligodeoxynucleotide CpG (CpG) on promoting homologous protection and cross-protection after vaccination of C57BL/6 and BALB/c mice with inactivated split virus. Combination adjuvant effects of MPL and CpG on improving homologous and cross protective vaccine efficacy were evident as shown by higher levels of homologous and cross-reactive binding IgG and hemagglutination inhibiting antibodies. Combination adjuvant effects on enhancing the protective efficacy against homologous and heterosubtypic virus were demonstrated by less weight loss, lower airway inflammatory disease, and better control of viral loads as well as prevention of inflammatory cytokines and cellular infiltrates. Overall, the findings in this study suggest that a combination adjuvant of different toll-like receptor ligands exhibits a unique pattern of innate and adaptive immune responses, contributing to improved homologous and heterosubtypic cross-protection by inactivated split virion influenza vaccination.
Collapse
Affiliation(s)
- Eun-Ju Ko
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Yu-Jin Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037 USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
33
|
Hoffmann FM, Berger JL, Lingel I, Laumonnier Y, Lewkowich IP, Schmudde I, König P. Distribution and Interaction of Murine Pulmonary Phagocytes in the Naive and Allergic Lung. Front Immunol 2018; 9:1046. [PMID: 29868009 PMCID: PMC5964136 DOI: 10.3389/fimmu.2018.01046] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 04/26/2018] [Indexed: 12/19/2022] Open
Abstract
The division of labor between pulmonary phagocytic subsets [macrophage/monocyte and dendritic cell (DC) subpopulations] has been described at the functional level. However, whether these lung phagocytes also display unique spatial distribution remains unclear. Here, to analyze cellular distribution in lung compartments and contacts between phagocyte subpopulations, we established an immunohistochemistry (IHC)-based method to clearly identify murine lung phagocyte subsets in situ based on differential expression of CD11c, CD11b, MHC-II, Langerin and mPDCA-1. Furthermore, we investigated subset-specific functional differences in antigen uptake and spatial changes upon allergic sensitization. Our staining allowed the distinction between alveolar macrophages (AMs), interstitial macrophage (IM) subpopulations, CD11b+ DC subpopulations, CD103+ DCs, and plasmacytoid DCs (pDCs). We identified interstitial regions between airways and around airways as regions of IM/CD11b+ DC/CD103+ DC clusters, where a subset of IMs (IM2) and CD103+ DCs formed intense contacts that decreased upon allergic sensitization. These data indicate functional interactions between both cell types either in steady state or after antigen encounter affecting the development of allergies or tolerance. Furthermore, we observed major antigen uptake in AMs and IMs rather than DC subpopulations that was not restricted to airways and adjacent areas. This will enable to focus future studies to immunologically relevant cellular interactions and to unravel which cells are tipping the balance between pro-inflammatory immune responses or tolerance.
Collapse
Affiliation(s)
- Franziska M Hoffmann
- Institute of Anatomy, University of Lübeck, Lübeck, Germany.,Airway Research Center North (ARCN), German Center for Lung Research (DZL), Lübeck, Germany
| | - Johann L Berger
- Institute of Anatomy, University of Lübeck, Lübeck, Germany.,Airway Research Center North (ARCN), German Center for Lung Research (DZL), Lübeck, Germany
| | - Imke Lingel
- Institute of Anatomy, University of Lübeck, Lübeck, Germany.,Airway Research Center North (ARCN), German Center for Lung Research (DZL), Lübeck, Germany
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Ian P Lewkowich
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| | - Inken Schmudde
- Institute of Anatomy, University of Lübeck, Lübeck, Germany.,Airway Research Center North (ARCN), German Center for Lung Research (DZL), Lübeck, Germany
| | - Peter König
- Institute of Anatomy, University of Lübeck, Lübeck, Germany.,Airway Research Center North (ARCN), German Center for Lung Research (DZL), Lübeck, Germany
| |
Collapse
|
34
|
Zhou Y, Horowitz JC, Naba A, Ambalavanan N, Atabai K, Balestrini J, Bitterman PB, Corley RA, Ding BS, Engler AJ, Hansen KC, Hagood JS, Kheradmand F, Lin QS, Neptune E, Niklason L, Ortiz LA, Parks WC, Tschumperlin DJ, White ES, Chapman HA, Thannickal VJ. Extracellular matrix in lung development, homeostasis and disease. Matrix Biol 2018. [PMID: 29524630 DOI: 10.1016/j.matbio.2018.03.005] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The lung's unique extracellular matrix (ECM), while providing structural support for cells, is critical in the regulation of developmental organogenesis, homeostasis and injury-repair responses. The ECM, via biochemical or biomechanical cues, regulates diverse cell functions, fate and phenotype. The composition and function of lung ECM become markedly deranged in pathological tissue remodeling. ECM-based therapeutics and bioengineering approaches represent promising novel strategies for regeneration/repair of the lung and treatment of chronic lung diseases. In this review, we assess the current state of lung ECM biology, including fundamental advances in ECM composition, dynamics, topography, and biomechanics; the role of the ECM in normal and aberrant lung development, adult lung diseases and autoimmunity; and ECM in the regulation of the stem cell niche. We identify opportunities to advance the field of lung ECM biology and provide a set recommendations for research priorities to advance knowledge that would inform novel approaches to the pathogenesis, diagnosis, and treatment of chronic lung diseases.
Collapse
Affiliation(s)
- Yong Zhou
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, United States.
| | - Jeffrey C Horowitz
- Division of Pulmonary and Critical Care Medicine, University of Michigan, United States.
| | - Alexandra Naba
- Department of Physiology & Biophysics, University of Illinois at Chicago, United States.
| | | | - Kamran Atabai
- Lung Biology Center, University of California, San Francisco, United States.
| | | | | | - Richard A Corley
- Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, United States.
| | - Bi-Sen Ding
- Weill Cornell Medical College, United States.
| | - Adam J Engler
- Sanford Consortium for Regenerative Medicine, University of California, San Diego, United States.
| | - Kirk C Hansen
- Biochemistry & Molecular Genetics, University of Colorado Denver, United States.
| | - James S Hagood
- Pediatric Respiratory Medicine, University of California San Diego, United States.
| | - Farrah Kheradmand
- Division of Pulmonary and Critical Care, Baylor College of Medicine, United States.
| | - Qing S Lin
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, United States.
| | - Enid Neptune
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, United States.
| | - Laura Niklason
- Department of Anesthesiology, Yale University, United States.
| | - Luis A Ortiz
- Division of Environmental and Occupational Health, University of Pittsburgh, United States.
| | - William C Parks
- Department of Medicine, Cedars-Sinai Medical Center, United States.
| | - Daniel J Tschumperlin
- Department of Physiology & Biomedical Engineering, Mayo Clinic College of Medicine, United States.
| | - Eric S White
- Division of Pulmonary and Critical Care Medicine, University of Michigan, United States.
| | - Harold A Chapman
- Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, United States.
| | - Victor J Thannickal
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, United States.
| |
Collapse
|
35
|
Chen K, Bao Z, Gong W, Tang P, Yoshimura T, Wang JM. Regulation of inflammation by members of the formyl-peptide receptor family. J Autoimmun 2017; 85:64-77. [PMID: 28689639 PMCID: PMC5705339 DOI: 10.1016/j.jaut.2017.06.012] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 06/29/2017] [Indexed: 12/14/2022]
Abstract
Inflammation is associated with a variety of diseases. The hallmark of inflammation is leukocyte infiltration at disease sites in response to pathogen- or damage-associated chemotactic molecular patterns (PAMPs and MAMPs), which are recognized by a superfamily of seven transmembrane, Gi-protein-coupled receptors (GPCRs) on cell surface. Chemotactic GPCRs are composed of two major subfamilies: the classical GPCRs and chemokine GPCRs. Formyl-peptide receptors (FPRs) belong to the classical chemotactic GPCR subfamily with unique properties that are increasingly appreciated for their expression on diverse host cell types and the capacity to interact with a plethora of chemotactic PAMPs and MAMPs. Three FPRs have been identified in human: FPR1-FPR3, with putative corresponding mouse counterparts. FPR expression was initially described in myeloid cells but subsequently in many non-hematopoietic cells including cancer cells. Accumulating evidence demonstrates that FPRs possess multiple functions in addition to controlling inflammation, and participate in the processes of many pathophysiologic conditions. They are not only critical mediators of myeloid cell trafficking, but are also implicated in tissue repair, angiogenesis and protection against inflammation-associated tumorigenesis. A series recent discoveries have greatly expanded the scope of FPRs in host defense which uncovered the essential participation of FPRs in step-wise trafficking of myeloid cells including neutrophils and dendritic cells (DCs) in host responses to bacterial infection, tissue injury and wound healing. Also of great interest is the FPRs are exploited by malignant cancer cells for their growth, invasion and metastasis. In this article, we review the current understanding of FPRs concerning their expression in a vast array of cell types, their involvement in guiding leukocyte trafficking in pathophysiological conditions, and their capacity to promote the differentiation of immune cells, their participation in tumor-associated inflammation and cancer progression. The close association of FPRs with human diseases and cancer indicates their potential as targets for the development of therapeutics.
Collapse
Affiliation(s)
- Keqiang Chen
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA
| | - Zhiyao Bao
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA; Department of Pulmonary & Critical Care Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| | - Peng Tang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA; Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Teizo Yoshimura
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Ji Ming Wang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA.
| |
Collapse
|
36
|
Baharom F, Rankin G, Blomberg A, Smed-Sörensen A. Human Lung Mononuclear Phagocytes in Health and Disease. Front Immunol 2017; 8:499. [PMID: 28507549 PMCID: PMC5410584 DOI: 10.3389/fimmu.2017.00499] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/11/2017] [Indexed: 12/17/2022] Open
Abstract
The lungs are vulnerable to attack by respiratory insults such as toxins, allergens, and pathogens, given their continuous exposure to the air we breathe. Our immune system has evolved to provide protection against an array of potential threats without causing collateral damage to the lung tissue. In order to swiftly detect invading pathogens, monocytes, macrophages, and dendritic cells (DCs)-together termed mononuclear phagocytes (MNPs)-line the respiratory tract with the key task of surveying the lung microenvironment in order to discriminate between harmless and harmful antigens and initiate immune responses when necessary. Each cell type excels at specific tasks: monocytes produce large amounts of cytokines, macrophages are highly phagocytic, whereas DCs excel at activating naïve T cells. Extensive studies in murine models have established a division of labor between the different populations of MNPs at steady state and during infection or inflammation. However, a translation of important findings in mice is only beginning to be explored in humans, given the challenge of working with rare cells in inaccessible human tissues. Important progress has been made in recent years on the phenotype and function of human lung MNPs. In addition to a substantial population of alveolar macrophages, three subsets of DCs have been identified in the human airways at steady state. More recently, monocyte-derived cells have also been described in healthy human lungs. Depending on the source of samples, such as lung tissue resections or bronchoalveolar lavage, the specific subsets of MNPs recovered may differ. This review provides an update on existing studies investigating human respiratory MNP populations during health and disease. Often, inflammatory MNPs are found to accumulate in the lungs of patients with pulmonary conditions. In respiratory infections or inflammatory diseases, this may contribute to disease severity, but in cancer patients this may improve clinical outcomes. By expanding on this knowledge, specific lung MNPs may be targeted or modulated in order to attain favorable responses that can improve preventive or treatment strategies against respiratory infections, lung cancer, or lung inflammatory diseases.
Collapse
Affiliation(s)
- Faezzah Baharom
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Gregory Rankin
- Department of Public Health and Clinical Medicine, Division of Medicine, Umeå University, Umeå, Sweden
| | - Anders Blomberg
- Department of Public Health and Clinical Medicine, Division of Medicine, Umeå University, Umeå, Sweden
| | - Anna Smed-Sörensen
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
37
|
Takamura S. Persistence in Temporary Lung Niches: A Survival Strategy of Lung-Resident Memory CD8 + T Cells. Viral Immunol 2017; 30:438-450. [PMID: 28418771 PMCID: PMC5512299 DOI: 10.1089/vim.2017.0016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Respiratory virus infections, such as those mediated by influenza virus, parainfluenza virus, respiratory syncytial virus (RSV), severe acute respiratory syndrome coronavirus (SARS-CoV), rhinovirus, and adenovirus, are responsible for substantial morbidity and mortality, especially in children and older adults. Furthermore, the potential emergence of highly pathogenic strains of influenza virus poses a significant public health threat. Thus, the development of vaccines capable of eliciting long-lasting protective immunity to those pathogens is a major public health priority. CD8+ Tissue-resident memory T (TRM) cells are a newly defined population that resides permanently in the nonlymphoid tissues including the lung. These cells are capable of providing local protection immediately after infection, thereby promoting rapid host recovery. Recent studies have offered new insights into the anatomical niches that harbor lung CD8+ TRM cells, and also identified the requirement and limitations of TRM maintenance. However, it remains controversial whether lung CD8+ TRM cells are continuously replenished by new cells from the circulation or permanently lodged in this site. A better understanding of how lung CD8+ TRM cells are generated and maintained and the tissue-specific factors that drive local TRM formation is required for optimal vaccine development. This review focuses on recent advance in our understanding of CD8+ TRM cell establishment and maintenance in the lung, and describes how those processes are uniquely regulated in this tissue.
Collapse
Affiliation(s)
- Shiki Takamura
- Department of Immunology, Kindai University , Faculty of Medicine, Osaka, Japan
| |
Collapse
|
38
|
Intubation-free in vivo imaging of the tracheal mucosa using two-photon microscopy. Sci Rep 2017; 7:694. [PMID: 28386104 PMCID: PMC5429620 DOI: 10.1038/s41598-017-00769-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 03/09/2017] [Indexed: 01/08/2023] Open
Abstract
The mucosal layer of conducting airways is the primary tissue exposed to inhaled microorganisms, allergens and pollutants. We developed an in vivo two-photon microscopic approach that allows performing dynamic imaging studies in the mouse trachea, which is a commonly used in vivo model of human small-diameter bronchi. By providing stabilized access to the tracheal mucosa without intubation, our setup uniquely allows dynamic in vivo imaging of mucociliary clearance and steady-state immune cell behavior within the complex airway mucosal tissue.
Collapse
|
39
|
de Leve S, Wirsdörfer F, Cappuccini F, Schütze A, Meyer AV, Röck K, Thompson LF, Fischer JW, Stuschke M, Jendrossek V. Loss of CD73 prevents accumulation of alternatively activated macrophages and the formation of prefibrotic macrophage clusters in irradiated lungs. FASEB J 2017; 31:2869-2880. [PMID: 28325757 DOI: 10.1096/fj.201601228r] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 03/06/2017] [Indexed: 12/17/2022]
Abstract
While radiotherapy is a mainstay for cancer therapy, pneumonitis and fibrosis constitute dose-limiting side effects of thorax and whole body irradiation. So far, the contribution of immune cells to disease progression is largely unknown. Here we studied the role of ecto-5'-nucelotidase (CD73)/adenosine-induced changes in the myeloid compartment in radiation-induced lung fibrosis. C57BL/6 wild-type or CD73-/- mice received a single dose of whole thorax irradiation (WTI, 15 Gy). Myeloid cells were characterized in flow cytometric, histologic, and immunohistochemical analyses as well as RNA analyses. WTI induced a pronounced reduction of alveolar macrophages in both strains that recovered within 6 wk. Fibrosis development in wild-type mice was associated with a time-dependent deposition of hyaluronic acid (HA) and increased expression of markers for alternative activation on alveolar macrophages. These include the antiinflammatory macrophage mannose receptor and arginase-1. Further, macrophages accumulated in organized clusters and expressed profibrotic mediators at ≥25 wk after irradiation (fibrotic phase). Irradiated CD73-/- mice showed an altered regulation of components of the HA system and no clusters of alternatively activated macrophages. We speculate that accumulation of alternatively activated macrophages in organized clusters represents the origins of fibrotic foci after WTI and is promoted by a cross-talk between HA, CD73/adenosine signaling, and other profibrotic mediators.-De Leve, S., Wirsdörfer, F., Cappuccini, F., Schütze, A., Meyer, A. V., Röck, K., Thompson, L. F., Fischer, J. W., Stuschke, M., Jendrossek, V. Loss of CD73 prevents accumulation of alternatively activated macrophages and the formation of prefibrotic macrophage clusters in irradiated lungs.
Collapse
Affiliation(s)
- Simone de Leve
- Institute of Cell Biology (Cancer Research), University Hospital Essen, Essen, Germany
| | - Florian Wirsdörfer
- Institute of Cell Biology (Cancer Research), University Hospital Essen, Essen, Germany
| | - Federica Cappuccini
- Institute of Cell Biology (Cancer Research), University Hospital Essen, Essen, Germany
| | - Alexandra Schütze
- Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Alina V Meyer
- Institute of Cell Biology (Cancer Research), University Hospital Essen, Essen, Germany
| | - Katharina Röck
- Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Linda F Thompson
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Jens W Fischer
- Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Martin Stuschke
- Department of Radiation Oncology, University Hospital Essen, Essen, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, Essen, Germany;
| |
Collapse
|
40
|
Cytokine regulation of lung Th17 response to airway immunization using LPS adjuvant. Mucosal Immunol 2017; 10:361-372. [PMID: 27328989 PMCID: PMC5179326 DOI: 10.1038/mi.2016.54] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 05/10/2016] [Indexed: 02/04/2023]
Abstract
Infections caused by bacteria in the airway preferentially induce a Th17 response. However, the mechanisms involved in the regulation of CD4 T-cell responses in the lungs are incompletely understood. Here, we have investigated the mechanisms involved in the regulation of Th17 differentiation in the lungs in response to immunization with lipopolysaccharide (LPS) as an adjuvant. Our data show that both Myd88 and TRIF are necessary for Th17 induction. This distinctive fate determination can be accounted for by the pattern of inflammatory cytokines induced by airway administration of LPS. We identified the production of interleukin (IL)-1β and IL-6 by small macrophages and IL-23 by alveolar dendritic cells (DCs), favoring Th17 responses, and IL-10 repressing interferon (IFN)-γ production. Furthermore, we show that exogenous IL-1β can drastically alter Th1 responses driven by influenza and lymphocytic choriomeningitis virus infection models and induce IL-17 production. Thus, the precision of the lung immune responses to potential threats is orchestrated by the cytokine microenvironment, can be repolarized and targeted therapeutically by altering the cytokine milieu. These results indicate that how the development of Th17 responses in the lung is regulated by the cytokines produced by lung DCs and macrophages in response to intranasal immunization with LPS adjuvant.
Collapse
|
41
|
Influenza and Memory T Cells: How to Awake the Force. Vaccines (Basel) 2016; 4:vaccines4040033. [PMID: 27754364 PMCID: PMC5192353 DOI: 10.3390/vaccines4040033] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 09/27/2016] [Indexed: 12/24/2022] Open
Abstract
Annual influenza vaccination is an effective way to prevent human influenza. Current vaccines are mainly focused on eliciting a strain-matched humoral immune response, requiring yearly updates, and do not provide protection for all vaccinated individuals. The past few years, the importance of cellular immunity, and especially memory T cells, in long-lived protection against influenza virus has become clear. To overcome the shortcomings of current influenza vaccines, eliciting both humoral and cellular immunity is imperative. Today, several new vaccines such as infection-permissive and recombinant T cell inducing vaccines, are being developed and show promising results. These vaccines will allow us to stay several steps ahead of the constantly evolving influenza virus.
Collapse
|
42
|
P2X7 receptor-dependent tuning of gut epithelial responses to infection. Immunol Cell Biol 2016; 95:178-188. [PMID: 27559003 PMCID: PMC5181772 DOI: 10.1038/icb.2016.75] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 08/16/2016] [Accepted: 08/20/2016] [Indexed: 12/23/2022]
Abstract
Infection and injury of the gut are associated with cell damage and release of molecules such as extracellular adenosine 5'-triphosphate (ATP), which is recognised by the purinergic P2X7 receptor (P2X7R). P2X7R is widely expressed in the gut by antigen-presenting cells (APCs) and epithelial cells, but the role of the P2X7R on epithelial cells is poorly understood. We investigated P2X7R in intestinal epithelium in vitro and in vivo using two model infections, Toxoplasma gondii and Trichinella spiralis. Lipopolysaccharide and ATP treatment of intestinal epithelial cells and infection with T. gondii in vitro did not promote inflammasome-associated interleukin-1β (IL-1β) or IL-18 secretion, but promoted C-C motif chemokine ligand 5 (CCL5), tumour necrosis factor-α and IL-6 production that were significantly reduced when the P2X7R was blocked. Similarly, in vivo, infection with either T. spiralis or T. gondii induced rapid upregulation of epithelial CCL5 in wild-type (wild-type (WT)) mice that was significantly reduced in P2X7R-/- littermate controls. The effects of reduced epithelial CCL5 were assayed by investigating recruitment of dendritic cells (DCs) to the epithelium. Infection induced a rapid recruitment of CD11c+CD103+ DC subsets into the epithelial layer of WT mice but not P2X7R-/- mice. In vitro chemotaxis assays and bone marrow chimeras demonstrated the importance of epithelial P2X7R in DC recruitment. P2X7R signalling in epithelial cells mediates chemokine responses to promote initiation of host immunity to infection.
Collapse
|
43
|
Schenk H, Neumann D, Kloth C. Histamine regulates murine primary dendritic cell functions. Immunopharmacol Immunotoxicol 2016; 38:379-84. [PMID: 27560832 DOI: 10.1080/08923973.2016.1214144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE AND DESIGN The modulation of antigen uptake and activation of dendritic cells (DCs) by histamine may function as a regulator of inflammation. Therefore, we sought to determine the impact of histamine on antigen uptake by and activation of murine DCs. MATERIAL AND METHODS DCs from spleen and lung were either identified by flow cytometry or were immunomagnetically enriched. Cells were stimulated with histamine, and the regulation of MHC-II and co-stimulatory molecule expression (CD80, CD86, and ICOS-L) and antigen uptake were quantified by flow cytometry. Individual contributions of the histamine receptor subtypes were determined by using the antagonists mepyramine (histamine H1-receptor: H1R), famotidine (H2R), and JNJ 7777120 (H4R). RESULTS Histamine accelerated the uptake of soluble antigen via the H1R, H2R, and H4R in splenic DCs. Co-stimulatory molecule expression was enhanced already by enrichment procedures, thus, the analyses were performed in unseparated cell populations. Histamine enhanced the expression of CD86 and ICOS-L while expression of CD80 was unaffected. Antagonism at H1R, H2R, and H4R and at H1R and H4R reduced the histamine-induced enhanced expression of CD86 and ICOS-L, respectively. CONCLUSIONS Histamine contributes to the regulation of the immunological synapse by stimulation of antigen uptake and activation of DCs via H1R, H2R, and H4R.
Collapse
Affiliation(s)
- Heiko Schenk
- a Institute of Pharmacology, Hannover Medical School , Hannover , Germany
| | - Detlef Neumann
- a Institute of Pharmacology, Hannover Medical School , Hannover , Germany
| | - Christina Kloth
- a Institute of Pharmacology, Hannover Medical School , Hannover , Germany ;,b Institute of Applied Anatomy, Hannover Medical School , Hannover , Germany
| |
Collapse
|
44
|
Esmagambetov IB, Alekseeva SV, Sayadyan KS, Shmarov MM. CURRENT APPROACHES TO UNIVERSAL VACCINE AGAINST INFLUENZA VIRUS. RUSSIAN JOURNAL OF INFECTION AND IMMUNITY 2016. [DOI: 10.15789/2220-7619-2016-2-117-132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
45
|
Papazian D, Würtzen PA, Hansen SWK. Polarized Airway Epithelial Models for Immunological Co-Culture Studies. Int Arch Allergy Immunol 2016; 170:1-21. [PMID: 27240620 DOI: 10.1159/000445833] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Epithelial cells line all cavities and surfaces throughout the body and play a substantial role in maintaining tissue homeostasis. Asthma and other atopic diseases are increasing worldwide and allergic disorders are hypothesized to be a consequence of a combination of dysregulation of the epithelial response towards environmental antigens and genetic susceptibility, resulting in inflammation and T cell-derived immune responses. In vivo animal models have long been used to study immune homeostasis of the airways but are limited by species restriction and lack of exposure to a natural environment of both potential allergens and microflora. Limitations of these models prompt a need to develop new human cell-based in vitro models. A variety of co-culture systems for modelling the respiratory epithelium exist and are available to the scientific community. The models have become increasingly sophisticated and specific care needs to be taken with regard to cell types, culture medium and culture models, depending on the aim of the study. Although great strides have been made, there is still a need for further optimization, and optimally also for standardization, in order for in vitro co-culture models to become powerful tools in the discovery of key molecules dictating immunity and/or tolerance, and for understanding the complex interplay that takes place between mucosa, airway epithelium and resident or infiltrating immune cells. This review focuses on current knowledge and the advantages and limitations of the different cell types and culture methods used in co-culture models of the human airways.
Collapse
Affiliation(s)
- Dick Papazian
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | | |
Collapse
|
46
|
Kini Bailur J, Gueckel B, Pawelec G. Prognostic impact of high levels of circulating plasmacytoid dendritic cells in breast cancer. J Transl Med 2016; 14:151. [PMID: 27234566 PMCID: PMC4884426 DOI: 10.1186/s12967-016-0905-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 05/13/2016] [Indexed: 12/24/2022] Open
Abstract
Background Identifying immune markers in blood that are informative for breast cancer patient survival would not only be useful for prognosis but might also provide mechanistic insights into processes facilitating survival. Methods We phenotyped circulating plasmacytoid dendritic cells (pDCs), myeloid-derived suppressor cells (MDSCs) and regulatory T-cells in relation to T-cell responses to Her-2 in vitro in 75 untreated breast cancer patients 28–87 years of age at diagnosis. Results Patients with later stage tumors had lower levels of circulating pDCs (p = 0.008). There was a positive association between 5-year survival and higher than median levels of circulating pDCs (p = 0.03). We confirmed that 5-year survival correlated with CD8+ but not CD4+ T-cell responsiveness to Her-2 peptides in this cohort of younger and older patients (p = 0.04). Including pDCs in the analysis of previously-established parameters revealed that patients who had a CD8+ T-cell response to Her-2 together with a low ratio of MDSCs:pDCs had 100 % 5-year survival. High levels of pDCs and the presence of a CD8+ T-cell response to Her-2 were independent positive survival indicators according to multivariate Cox analysis. Conclusions Our new results suggest that circulating pDCs could be a positive prognostic indicator in breast cancer patients of all ages, together with the previously established CD8+ T-cell reactivity to Her-2 antigens in older patients only. These two prognostic indicators were independent and emphasize the important role of immunity in ensuring breast cancer patient survival, even in those not undergoing immunotherapy. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0905-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jithendra Kini Bailur
- Department of Internal Medicine II, Centre for Medical Research, University Hospital Tübingen, Waldhoernlestr. 22, 72072, Tübingen, Germany. .,Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
| | - Brigitte Gueckel
- Radiology Clinic, Diagnostic and Interventional Radiology, University Hospital Tübingen, Tübingen, Germany
| | - Graham Pawelec
- Department of Internal Medicine II, Centre for Medical Research, University Hospital Tübingen, Waldhoernlestr. 22, 72072, Tübingen, Germany.,School of Science and Technology, College of Arts and Science, Nottingham Trent University, Nottingham, UK.,Division of Cancer Studies, Faculty of Life Sciences and Medicine, King's College London, London, UK.,Institute of Cancer Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
47
|
Chen K, Wang JM, Yuan R, Yi X, Li L, Gong W, Yang T, Li L, Su S. Tissue-resident dendritic cells and diseases involving dendritic cell malfunction. Int Immunopharmacol 2016; 34:1-15. [PMID: 26906720 PMCID: PMC4818737 DOI: 10.1016/j.intimp.2016.02.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/05/2016] [Indexed: 01/10/2023]
Abstract
Dendritic cells (DCs) control immune responses and are central to the development of immune memory and tolerance. DCs initiate and orchestrate immune responses in a manner that depends on signals they receive from microbes and cellular environment. Although DCs consist mainly of bone marrow-derived and resident populations, a third tissue-derived population resides the spleen and lymph nodes (LNs), different subsets of tissue-derived DCs have been identified in the blood, spleen, lymph nodes, skin, lung, liver, gut and kidney to maintain the tolerance and control immune responses. Tissue-resident DCs express different receptors for microbe-associated molecular patterns (MAMPs) and damage-associated molecular patterns (DAMPs), which were activated to promote the production of pro- or anti-inflammatory cytokines. Malfunction of DCs contributes to diseases such as autoimmunity, allergy, and cancer. It is therefore important to update the knowledge about resident DC subsets and diseases associated with DC malfunction.
Collapse
Affiliation(s)
- Keqiang Chen
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA; Laboratory of Inflammation Biology, Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061-0910, USA.
| | - Ji Ming Wang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA.
| | - Ruoxi Yuan
- Laboratory of Inflammation Biology, Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061-0910, USA
| | - Xiang Yi
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Liangzhu Li
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Wanghua Gong
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA; Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Tianshu Yang
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Liwu Li
- Laboratory of Inflammation Biology, Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061-0910, USA
| | - Shaobo Su
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
48
|
Hoffmann F, Ender F, Schmudde I, Lewkowich IP, Köhl J, König P, Laumonnier Y. Origin, Localization, and Immunoregulatory Properties of Pulmonary Phagocytes in Allergic Asthma. Front Immunol 2016; 7:107. [PMID: 27047494 PMCID: PMC4803735 DOI: 10.3389/fimmu.2016.00107] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 03/08/2016] [Indexed: 01/21/2023] Open
Abstract
Allergic asthma is a chronic inflammatory disease of the airways that is driven by maladaptive T helper 2 (Th2) and Th17 immune responses against harmless, airborne substances. Pulmonary phagocytes represent the first line of defense in the lung where they constantly sense the local environment for potential threats. They comprise two distinct cell types, i.e., macrophages and dendritic cells (DC) that differ in their origins and functions. Alveolar macrophages quickly take up most of the inhaled allergens, yet do not deliver their cargo to naive T cells sampling in draining lymph nodes. In contrast, pulmonary DCs instruct CD4(+) T cells develop into Th2 and Th17 effectors, initiating the maladaptive immune responses toward harmless environmental substances observed in allergic individuals. Unraveling the mechanisms underlying this mistaken identity of harmless, airborne substances by innate immune cells is one of the great challenges in asthma research. The identification of different pulmonary DC subsets, their role in antigen uptake, migration to the draining lymph nodes, and their potential to instruct distinct T cell responses has set the stage to unravel this mystery. However, at this point, a detailed understanding of the spatiotemporal resolution of DC subset localization, allergen uptake, processing, autocrine and paracrine cellular crosstalk, and the humoral factors that define the activation status of DCs is still lacking. In addition to DCs, at least two distinct macrophage populations have been identified in the lung that are either located in the airway/alveolar lumen or in the interstitium. Recent data suggest that such populations can exert either pro- or anti-inflammatory functions. Similar to the DC subsets, detailed insights into the individual roles of alveolar and interstitial macrophages during the different phases of asthma development are still missing. Here, we will provide an update on the current understanding of the origin, localization, and function of the diverse pulmonary antigen-presenting cell subsets, in particular with regard to the development and regulation of allergic asthma. While most data are from mouse models of experimental asthma, we have also included available human data to judge the translational value of the findings obtained in experimental asthma models.
Collapse
Affiliation(s)
| | - Fanny Ender
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Inken Schmudde
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Ian P. Lewkowich
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Giessen, Germany
| | - Peter König
- Institute for Anatomy, University of Lübeck, Lübeck, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Giessen, Germany
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| |
Collapse
|
49
|
Weitnauer M, Mijošek V, Dalpke AH. Control of local immunity by airway epithelial cells. Mucosal Immunol 2016; 9:287-98. [PMID: 26627458 DOI: 10.1038/mi.2015.126] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 10/25/2015] [Indexed: 02/04/2023]
Abstract
The lung is ventilated by thousand liters of air per day. Inevitably, the respiratory system comes into contact with airborne microbial compounds, most of them harmless contaminants. Airway epithelial cells are known to have innate sensor functions, thus being able to detect microbial danger. To avoid chronic inflammation, the pulmonary system has developed specific means to control local immune responses. Even though airway epithelial cells can act as proinflammatory promoters, we propose that under homeostatic conditions airway epithelial cells are important modulators of immune responses in the lung. In this review, we discuss epithelial cell regulatory functions that control reactivity of professional immune cells within the microenvironment of the airways and how these mechanisms are altered in pulmonary diseases. Regulation by epithelial cells can be divided into two mechanisms: (1) mediators regulate epithelial cells' innate sensitivity in cis and (2) factors are produced that limit reactivity of immune cells in trans.
Collapse
Affiliation(s)
- M Weitnauer
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, Heidelberg, Germany
| | - V Mijošek
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, Heidelberg, Germany
| | - A H Dalpke
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, Heidelberg, Germany.,Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
50
|
Smith N, Herbeuval JP. [Plasmacytoid dendritic cells: the novel Eldorado for antiviral therapy?]. Biol Aujourdhui 2015; 209:135-44. [PMID: 26514383 DOI: 10.1051/jbio/2015017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Indexed: 12/14/2022]
Abstract
Plasmacytoid dendritic cells (pDCs) represent the first line of host defense against viruses and are an essential link between innate and adaptive immunity. The antiviral factor IFN-α is massively produced by pDCs in response to HIV infection and induces the expression of cellular genes that interfere with viral replication (ISG). Indeed, type I IFN produced by pDCs has a direct anti-viral activity against HIV and has important adjuvant function on other immune cell-types, such as T cells, macrophages and dendritic cells. However, the role of type I IFN in HIV disease is complex and may depend on the stage of the disease. The immunologic hallmark of HIV infection is a status of chronic and progressive immune activation, which drives the immune system to exhaustion and leads to severe immunodeficiency. There is now strong evidence that chronic activation of pDCs may promote HIV pathogenesis and have an impact on adaptive T-cell response. Thus, targeting pDCs and type I IFN may open new therapeutic strategies for chronically activated HIV patients.
Collapse
Affiliation(s)
- Nikaïa Smith
- Equipe Chimie et Biologie, Nucléo(s)tides & Immunologie pour la Thérapie (CBNIT), CNRS UMR8601, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, CICB-Paris (FR 3567), Centre Universitaire des Saints-Pères, 45 rue des Saints Pères, 75006 Paris, France - Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jean-Philippe Herbeuval
- Equipe Chimie et Biologie, Nucléo(s)tides & Immunologie pour la Thérapie (CBNIT), CNRS UMR8601, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, CICB-Paris (FR 3567), Centre Universitaire des Saints-Pères, 45 rue des Saints Pères, 75006 Paris, France - Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|