1
|
Mandanas MV, Barrett NA. Epithelial sensing in allergic disease. Curr Opin Immunol 2024; 91:102490. [PMID: 39326203 PMCID: PMC11609016 DOI: 10.1016/j.coi.2024.102490] [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: 06/10/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024]
Abstract
Epithelial cells provide a first line of immune defense by maintaining barrier function, orchestrating mucociliary clearance, secreting antimicrobial molecules, and generating sentinel signals to both activate innate immune cells and shape adaptive immunity. Although epithelial alarmins play a particularly important role in the initiation of type 2 inflammation in response to allergens, the mechanisms by which epithelial cells sense the environment and regulate the generation and release of alarmins have been poorly understood. Recent studies have identified new sensors and signaling pathways used by barrier epithelial cells to elicit type 2 inflammation, including a novel pathway for the release of interleukin-33 from the nucleus that depends on apoptotic signaling. These recent findings have implications in the development of allergic diseases, from atopic eczema to food allergy, rhinitis, and asthma.
Collapse
Affiliation(s)
- Michael V Mandanas
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, MA, USA; Department of Immunology, Harvard Medical School, MA, USA
| | - Nora A Barrett
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, MA, USA; Department of Medicine, Harvard Medical School, MA, USA.
| |
Collapse
|
2
|
Lopez Espinoza A, Christopher T, Tait Wojno ED. Epithelial-immune interactions govern type 2 immunity at barrier surfaces. Curr Opin Immunol 2024; 91:102501. [PMID: 39522453 DOI: 10.1016/j.coi.2024.102501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
Allergic diseases are acute and chronic inflammatory conditions resulting from disproportionate responses to environmental stimuli. Affecting approximately 40% of the global population, these diseases significantly contribute to morbidity and increasing health care costs. Allergic reactions are triggered by pollen, house dust mites, animal dander, mold, food antigens, venoms, toxins, and drugs. This review explores the pivotal role of the epithelium in the skin, lungs, and gastrointestinal tract in regulating the allergic response and delves into the mechanisms of tissue-specific epithelial-immune interactions in this context, with recent advances highlighting their roles in the initiation, elicitation, and resolution phases of allergy. Understanding these intricate interactions at epithelial barriers is essential for developing targeted therapies to manage and treat allergic diseases.
Collapse
Affiliation(s)
| | - Tighe Christopher
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Elia D Tait Wojno
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
3
|
Saito H, Tamari M, Motomura K, Ikutani M, Nakae S, Matsumoto K, Morita H. Omics in allergy and asthma. J Allergy Clin Immunol 2024; 154:1378-1390. [PMID: 39384073 DOI: 10.1016/j.jaci.2024.09.023] [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: 09/09/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/11/2024]
Abstract
This review explores the transformative impact of omics technologies on allergy and asthma research in recent years, focusing on advancements in high-throughput technologies related to genomics and transcriptomics. In particular, the rapid spread of single-cell RNA sequencing has markedly advanced our understanding of the molecular pathology of allergic diseases. Furthermore, high-throughput genome sequencing has accelerated the discovery of monogenic disorders that were previously overlooked as ordinary intractable allergic diseases. We also introduce microbiomics, proteomics, lipidomics, and metabolomics, which are quickly growing areas of research interest, although many of their current findings remain inconclusive as solid evidence. By integrating these omics data, we will gain deeper insights into disease mechanisms, leading to the development of precision medicine approaches that promise to enhance treatment outcomes.
Collapse
Affiliation(s)
- Hirohisa Saito
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Masato Tamari
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kenichiro Motomura
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Masashi Ikutani
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Susumu Nakae
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan; Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Hideaki Morita
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; Allergy Center, National Center for Child Health and Development, Tokyo, Japan.
| |
Collapse
|
4
|
Brightling CE, Marone G, Aegerter H, Chanez P, Heffler E, Pavord ID, Rabe KF, Uller L, Dorscheid D. The epithelial era of asthma research: knowledge gaps and future direction for patient care. Eur Respir Rev 2024; 33:240221. [PMID: 39694589 DOI: 10.1183/16000617.0221-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/11/2024] [Indexed: 12/20/2024] Open
Abstract
The Epithelial Science Expert Group convened on 18-19 October 2023, in Naples, Italy, to discuss the current understanding of the fundamental role of the airway epithelium in asthma and other respiratory diseases and to explore the future direction of patient care. This review summarises the key concepts and research questions that were raised. As an introduction to the epithelial era of research, the evolution of asthma management throughout the ages was discussed and the role of the epithelium as an immune-functioning organ was elucidated. The role of the bronchial epithelial cells in lower airway diseases beyond severe asthma was considered, as well as the role of the epithelium in upper airway diseases such as chronic rhinosinusitis. The biology and application of biomarkers in patient care was also discussed. The Epithelial Science Expert Group also explored future research needs by identifying the current knowledge and research gaps in asthma management and ranking them by priority. It was identified that there is a need to define and support early assessment of asthma to characterise patients at high risk of severe asthma. Furthermore, a better understanding of asthma progression is required. The development of new treatments and diagnostic tests as well as the identification of new biomarkers will also be required to address the current unmet needs. Finally, an increased understanding of epithelial dysfunction will determine if we can alter disease progression and achieve clinical remission.
Collapse
Affiliation(s)
- Christopher E Brightling
- Institute for Lung Health, National Institute for Health and Care Research Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
- Joint first authors
| | - Gianni Marone
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research, School of Medicine, University of Naples Federico II, Naples, Italy
- Institute of Experimental Endocrinology and Oncology, National Research Council of Italy, Naples, Italy
- Joint first authors
| | - Helena Aegerter
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Pascal Chanez
- Department of Respiratory Diseases, Aix-Marseille University, Marseille, France
| | - Enrico Heffler
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Ian D Pavord
- Respiratory Medicine, National Institute for Health and Care Research Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Klaus F Rabe
- LungenClinic Grosshansdorf, Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
- Chirstian-Alrechts University Kiel, Member of the German Center for Lung Research (DZL), Kiel, Germany
| | - Lena Uller
- Unit of Respiratory Immunopharmacology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Del Dorscheid
- Center for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
5
|
Roe K. The epithelial cell types and their multi-phased defenses against fungi and other pathogens. Clin Chim Acta 2024; 563:119889. [PMID: 39117034 DOI: 10.1016/j.cca.2024.119889] [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: 07/02/2024] [Revised: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024]
Abstract
Mucus and its movements are essential to epithelial tissue immune defenses against pathogens, including fungal pathogens, which can infect respiratory, gastrointestinal or the genito-urinary tracts. Several epithelial cell types contribute to their immune defense. This review focuses on the respiratory tract because of its paramount importance, but the observations will apply to epithelial cell defenses of other mucosal tissue, including the gastrointestinal and genito-urinary tracts. Mucus and its movements can enhance or degrade the immune defenses of the respiratory tract, particularly the lungs. The enhancements include inhaled pathogen entrapments, including fungal pathogens, pollutants and particulates, for their removal. The detriments include smaller lung airway obstructions by mucus, impairing the physical removal of pathogens and impairing vital transfers of oxygen and carbon dioxide between the alveolar circulatory system and the pulmonary air. Inflammation, edema and/or alveolar cellular damage can also reduce vital transfers of oxygen and carbon dioxide between the lung alveolar circulatory system and the pulmonary air. Furthermore, respiratory tract defenses are affected by several fatty acid mediators which activate cellular receptors to manipulate neutrophils, macrophages, dendritic cells, various innate lymphoid cells including the natural killer cells, T cells, γδ T cells, mucosal-associated invariant T cells, NKT cells and mast cells. These mediators include the inflammatory and frequently immunosuppressive prostaglandins and leukotrienes, and the special pro-resolving mediators, which normally resolve inflammation and immunosuppression. The total effects on the various epithelial cell and immune cell types, after exposures to pathogens, pollutants or particulates, will determine respiratory tract health or disease.
Collapse
Affiliation(s)
- Kevin Roe
- Retired United States Patent and Trademark Office, San Jose, CA, United States.
| |
Collapse
|
6
|
Qin M, Fang Y, Zheng Q, Peng M, Wang L, Sang X, Cao G. Tissue microenvironment induces tissue specificity of ILC2. Cell Death Discov 2024; 10:324. [PMID: 39013890 PMCID: PMC11252336 DOI: 10.1038/s41420-024-02096-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/18/2024] Open
Abstract
Type 2 innate lymphoid cells were found to be members of the innate immune cell family, which is involved in innate and adaptive immunity to resist the invasion of foreign antigens and induce allergic reactions caused by allergens. The advancement of ILC2 research has pointed out that ILC2s have a high degree of diversity, challenging the notion of their homogeneity as a cellular population. An increasing number of studies indicate that ILC2 is a cell population with tissue specificity which can be induced by the tissue microenvironment. In addition, crosstalk between tissues can change ILC2 functions of migration and activation. Here, we emphasize that ILC2 undergoes adaptive changes under the regulation of the tissue microenvironment and distant tissues, thereby coordinating the organization's operation. In addition, ILC2 alterations induced by the tissue microenvironment are not limited to the ILC2 cell population, and ILC2 can also transdifferentiate into another class of ILC cell population (ILC1 or ILC3). In this review, we summarized the tissue-specific effects of ILC2 by tissue microenvironment and focused on the function of ILC2 in inter-tissue crosstalk. Lastly, we discussed the transdifferentiations of ILC2 caused by the abnormal change in tissue environment.
Collapse
Affiliation(s)
- Minjing Qin
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuanyuan Fang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qitong Zheng
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengyun Peng
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xia'nan Sang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
7
|
Conway AE, White AA, Borish L, Shaker M, Lee MW. Eicosanoids, innate immunity, and alarmins in aspirin-exacerbated respiratory disease. Ann Allergy Asthma Immunol 2024; 133:13-15. [PMID: 38432553 DOI: 10.1016/j.anai.2024.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Affiliation(s)
| | - Andrew A White
- Division of Allergy, Asthma, and Immunology, Scripps Clinic, San Diego, California
| | - Larry Borish
- Departments of Medicine and Microbiology, Asthma and Allergic Disease Center, University of Virginia Health System, Charlottesville, Virginia
| | - Marcus Shaker
- Departments of Medicine and Pediatrics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire; Section of Allergy and Clinical Immunology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire.
| | - Michael W Lee
- Department of Medical Education, Dartmouth Geisel School of Medicine, Hanover, New Hampshire
| |
Collapse
|
8
|
Alhallak K, Nagai J, Zaleski K, Marshall S, Salloum T, Derakhshan T, Hayashi H, Feng C, Kratchmarov R, Lai J, Kuchibhotla V, Nishida A, Balestrieri B, Laidlaw T, Dwyer DF, Boyce JA. Mast cells control lung type 2 inflammation via prostaglandin E 2-driven soluble ST2. Immunity 2024; 57:1274-1288.e6. [PMID: 38821053 PMCID: PMC11168874 DOI: 10.1016/j.immuni.2024.05.003] [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: 06/28/2023] [Revised: 01/26/2024] [Accepted: 05/06/2024] [Indexed: 06/02/2024]
Abstract
Severe asthma and sinus disease are consequences of type 2 inflammation (T2I), mediated by interleukin (IL)-33 signaling through its membrane-bound receptor, ST2. Soluble (s)ST2 reduces available IL-33 and limits T2I, but little is known about its regulation. We demonstrate that prostaglandin E2 (PGE2) drives production of sST2 to limit features of lung T2I. PGE2-deficient mice display diminished sST2. In humans with severe respiratory T2I, urinary PGE2 metabolites correlate with serum sST2. In mice, PGE2 enhanced sST2 secretion by mast cells (MCs). Mice lacking MCs, ST2 expression by MCs, or E prostanoid (EP)2 receptors by MCs showed reduced sST2 lung concentrations and strong T2I. Recombinant sST2 reduced T2I in mice lacking PGE2 or ST2 expression by MCs back to control levels. PGE2 deficiency also reversed the hyperinflammatory phenotype in mice lacking ST2 expression by MCs. PGE2 thus suppresses T2I through MC-derived sST2, explaining the severe T2I observed in low PGE2 states.
Collapse
Affiliation(s)
- Kinan Alhallak
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Jun Nagai
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Kendall Zaleski
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Sofia Marshall
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Tamara Salloum
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Tahereh Derakhshan
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Hiroaki Hayashi
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Chunli Feng
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Radomir Kratchmarov
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Juying Lai
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Virinchi Kuchibhotla
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Airi Nishida
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Barbara Balestrieri
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Tanya Laidlaw
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Daniel F Dwyer
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Joshua A Boyce
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
9
|
Li YH, Yang YS, Xue YB, Lei H, Zhang SS, Qian J, Yao Y, Zhou R, Huang L. G protein subunit G γ13-mediated signaling pathway is critical to the inflammation resolution and functional recovery of severely injured lungs. eLife 2024; 12:RP92956. [PMID: 38836551 DOI: 10.7554/elife.92956] [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] [Indexed: 06/06/2024] Open
Abstract
Tuft cells are a group of rare epithelial cells that can detect pathogenic microbes and parasites. Many of these cells express signaling proteins initially found in taste buds. It is, however, not well understood how these taste signaling proteins contribute to the response to the invading pathogens or to the recovery of injured tissues. In this study, we conditionally nullified the signaling G protein subunit Gγ13 and found that the number of ectopic tuft cells in the injured lung was reduced following the infection of the influenza virus H1N1. Furthermore, the infected mutant mice exhibited significantly larger areas of lung injury, increased macrophage infiltration, severer pulmonary epithelial leakage, augmented pyroptosis and cell death, greater bodyweight loss, slower recovery, worsened fibrosis and increased fatality. Our data demonstrate that the Gγ13-mediated signal transduction pathway is critical to tuft cells-mediated inflammation resolution and functional repair of the damaged lungs.To our best knowledge, it is the first report indicating subtype-specific contributions of tuft cells to the resolution and recovery.
Collapse
Affiliation(s)
- Yi-Hong Li
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Yi-Sen Yang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Yan-Bo Xue
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Hao Lei
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Sai-Sai Zhang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Junbin Qian
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Yushi Yao
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruhong Zhou
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Zhejiang University Shanghai Institute for Advanced Study, Shanghai, Shanghai, China
| | - Liquan Huang
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Zhejiang University Shanghai Institute for Advanced Study, Shanghai, Shanghai, China
- Monell Chemical Senses Center, Philadelphia, United States
| |
Collapse
|
10
|
Sipos F, Műzes G. Colonic Tuft Cells: The Less-Recognized Therapeutic Targets in Inflammatory Bowel Disease and Colorectal Cancer. Int J Mol Sci 2024; 25:6209. [PMID: 38892399 PMCID: PMC11172904 DOI: 10.3390/ijms25116209] [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: 04/28/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Tuft cells are more than guardian chemosensory elements of the digestive tract. They produce a variety of immunological effector molecules in response to stimulation; moreover, they are essential for defense against protozoa and nematodes. Beyond the description of their characteristics, this review aims to elucidate the potential pathogenic and therapeutic roles of colonic tuft cells in inflammatory bowel disease and colorectal cancer, focusing on their primarily immunomodulatory action. Regarding inflammatory bowel disease, tuft cells are implicated in both maintaining the integrity of the intestinal epithelial barrier and in tissue repair and regeneration processes. In addition to maintaining intestinal homeostasis, they display complex immune-regulatory functions. During the development of colorectal cancer, tuft cells can promote the epithelial-to-mesenchymal transition, alter the gastrointestinal microenvironment, and modulate both the anti-tumor immune response and the tumor microenvironment. A wide variety of their biological functions can be targeted for anti-inflammatory or anti-tumor therapies; however, the adverse side effects of immunomodulatory actions must be strictly considered.
Collapse
Affiliation(s)
- Ferenc Sipos
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| | - Györgyi Műzes
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| |
Collapse
|
11
|
Polverino F, Sin DD. Type 2 airway inflammation in COPD. Eur Respir J 2024; 63:2400150. [PMID: 38485148 DOI: 10.1183/13993003.00150-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/07/2024] [Indexed: 05/30/2024]
Abstract
Globally, nearly 400 million persons have COPD, and COPD is one of the leading causes of hospitalisation and mortality across the world. While it has been long-recognised that COPD is an inflammatory lung disease, dissimilar to asthma, type 2 inflammation was thought to play a minor role. However, recent studies suggest that in approximately one third of patients with COPD, type 2 inflammation may be an important driver of disease and a potential therapeutic target. Importantly, the immune cells and molecules involved in COPD-related type 2 immunity may be significantly different from those observed in severe asthma. Here, we identify the important molecules and effector immune cells involved in type 2 airway inflammation in COPD, discuss the recent therapeutic trial results of biologicals that have targeted these pathways and explore the future of therapeutic development of type 2 immune modulators in COPD.
Collapse
Affiliation(s)
- Francesca Polverino
- Pulmonary and Critical Care Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Don D Sin
- Centre for Heart Lung Innovation, St. Paul's Hospital and University of British Columbia Division of Respiratory Medicine, Vancouver, BC, Canada
| |
Collapse
|
12
|
Emanuel E, Arifuzzaman M, Artis D. Epithelial-neuronal-immune cell interactions: Implications for immunity, inflammation, and tissue homeostasis at mucosal sites. J Allergy Clin Immunol 2024; 153:1169-1180. [PMID: 38369030 PMCID: PMC11070312 DOI: 10.1016/j.jaci.2024.02.004] [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: 01/23/2024] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/20/2024]
Abstract
The epithelial lining of the respiratory tract and intestine provides a critical physical barrier to protect host tissues against environmental insults, including dietary antigens, allergens, chemicals, and microorganisms. In addition, specialized epithelial cells communicate directly with hematopoietic and neuronal cells. These epithelial-immune and epithelial-neuronal interactions control host immune responses and have important implications for inflammatory conditions associated with defects in the epithelial barrier, including asthma, allergy, and inflammatory bowel diseases. In this review, we discuss emerging research that identifies the mechanisms and impact of epithelial-immune and epithelial-neuronal cross talk in regulating immunity, inflammation, and tissue homeostasis at mucosal barrier surfaces. Understanding the regulation and impact of these pathways could provide new therapeutic targets for inflammatory diseases at mucosal sites.
Collapse
Affiliation(s)
- Elizabeth Emanuel
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY; Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY
| | - Mohammad Arifuzzaman
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY; Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY; Friedman Center for Nutrition and Inflammation, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY; Allen Discovery Center for Neuroimmune Interactions, New York, NY; Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY.
| |
Collapse
|
13
|
O'Carroll SM, Henkel FDR, O'Neill LAJ. Metabolic regulation of type I interferon production. Immunol Rev 2024; 323:276-287. [PMID: 38465724 DOI: 10.1111/imr.13318] [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] [Indexed: 03/12/2024]
Abstract
Over the past decade, there has been a surge in discoveries of how metabolic pathways regulate immune cell function in health and disease, establishing the field of immunometabolism. Specifically, pathways such as glycolysis, the tricarboxylic acid (TCA) cycle, and those involving lipid metabolism have been implicated in regulating immune cell function. Viral infections cause immunometabolic changes which lead to antiviral immunity, but little is known about how metabolic changes regulate interferon responses. Interferons are critical cytokines in host defense, rapidly induced upon pathogen recognition, but are also involved in autoimmune diseases. This review summarizes how metabolic change impacts interferon production. We describe how glycolysis, lipid metabolism (specifically involving eicosanoids and cholesterol), and the TCA cycle-linked intermediates itaconate and fumarate impact type I interferons. Targeting these metabolic changes presents new therapeutic possibilities to modulate type I interferons during host defense or autoimmune disorders.
Collapse
Affiliation(s)
- Shane M O'Carroll
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Fiona D R Henkel
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
14
|
Xu X, Yin J, Yang Y, Liu H, Yu J, Luo X, Zhang Y, Song X. Advances in co-pathogenesis of the united airway diseases. Respir Med 2024; 225:107580. [PMID: 38484897 DOI: 10.1016/j.rmed.2024.107580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/02/2024] [Accepted: 02/22/2024] [Indexed: 03/19/2024]
Abstract
According to the concept of "united airway diseases", the airway is a single organ in which upper and lower airway diseases are commonly comorbid. A range of inflammatory factors have been found to play an important role in the chain reaction of upper and lower airway diseases. However, the amount of research on this concept remains limited. The underlying mechanism of the relationship between typical diseases of the united airway, such as asthma, allergic rhinitis, and chronic sinusitis, also needs to be further explored. This review highlights the interaction between upper and lower respiratory diseases gathered from epidemiological, histoembryology, neural mechanistic, microbiological, and clinical studies, revealing the relationship between the upper and lower respiratory tracts.
Collapse
Affiliation(s)
- Xinjun Xu
- Department of Otolaryngology, Head and Neck Surgery. Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, China
| | - Jiali Yin
- Department of Otolaryngology, Head and Neck Surgery. Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, China
| | - Yujuan Yang
- Department of Otolaryngology, Head and Neck Surgery. Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, China
| | - Huifang Liu
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, China; The 2nd School of Clinical Medicine of Binzhou Medical University, Yantai, Shandong, China
| | - Jingyi Yu
- Department of Otolaryngology, Head and Neck Surgery. Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, China
| | - Xianghuang Luo
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, China; School of Clinical Medicine, Weifang Medical University, Weifang, 261042, China
| | - Yu Zhang
- Department of Otolaryngology, Head and Neck Surgery. Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, China.
| | - Xicheng Song
- Department of Otolaryngology, Head and Neck Surgery. Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, China.
| |
Collapse
|
15
|
Eshleman EM, Rice T, Potter C, Waddell A, Hashimoto-Hill S, Woo V, Field S, Engleman L, Lim HW, Schumacher MA, Frey MR, Denson LA, Finkelman FD, Alenghat T. Microbiota-derived butyrate restricts tuft cell differentiation via histone deacetylase 3 to modulate intestinal type 2 immunity. Immunity 2024; 57:319-332.e6. [PMID: 38295798 PMCID: PMC10901458 DOI: 10.1016/j.immuni.2024.01.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 10/14/2023] [Accepted: 01/05/2024] [Indexed: 02/03/2024]
Abstract
Tuft cells in mucosal tissues are key regulators of type 2 immunity. Here, we examined the impact of the microbiota on tuft cell biology in the intestine. Succinate induction of tuft cells and type 2 innate lymphoid cells was elevated with loss of gut microbiota. Colonization with butyrate-producing bacteria or treatment with butyrate suppressed this effect and reduced intestinal histone deacetylase activity. Epithelial-intrinsic deletion of the epigenetic-modifying enzyme histone deacetylase 3 (HDAC3) inhibited tuft cell expansion in vivo and impaired type 2 immune responses during helminth infection. Butyrate restricted stem cell differentiation into tuft cells, and inhibition of HDAC3 in adult mice and human intestinal organoids blocked tuft cell expansion. Collectively, these data define a HDAC3 mechanism in stem cells for tuft cell differentiation that is dampened by a commensal metabolite, revealing a pathway whereby the microbiota calibrate intestinal type 2 immunity.
Collapse
Affiliation(s)
- Emily M Eshleman
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Taylor Rice
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Crystal Potter
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Division of Immunology, Allergy and Rheumatology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Amanda Waddell
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Seika Hashimoto-Hill
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Vivienne Woo
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sydney Field
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Laura Engleman
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Hee-Woong Lim
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael A Schumacher
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA; Department of Pediatrics and Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mark R Frey
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA; Department of Pediatrics and Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lee A Denson
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Fred D Finkelman
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Division of Immunology, Allergy and Rheumatology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Theresa Alenghat
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
16
|
Ualiyeva S, Lemire E, Wong C, Perniss A, Boyd A, Avilés EC, Minichetti DG, Maxfield A, Roditi R, Matsumoto I, Wang X, Deng W, Barrett NA, Buchheit KM, Laidlaw TM, Boyce JA, Bankova LG, Haber AL. A nasal cell atlas reveals heterogeneity of tuft cells and their role in directing olfactory stem cell proliferation. Sci Immunol 2024; 9:eabq4341. [PMID: 38306414 PMCID: PMC11127180 DOI: 10.1126/sciimmunol.abq4341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 12/08/2023] [Indexed: 02/04/2024]
Abstract
The olfactory neuroepithelium serves as a sensory organ for odors and forms part of the nasal mucosal barrier. Olfactory sensory neurons are surrounded and supported by epithelial cells. Among them, microvillous cells (MVCs) are strategically positioned at the apical surface, but their specific functions are enigmatic, and their relationship to the other specialized epithelial cells is unclear. Here, we establish that the family of MVCs comprises tuft cells and ionocytes in both mice and humans. Integrating analysis of the respiratory and olfactory epithelia, we define the distinct receptor expression of TRPM5+ tuft-MVCs compared with Gɑ-gustducinhigh respiratory tuft cells and characterize a previously undescribed population of glandular DCLK1+ tuft cells. To establish how allergen sensing by tuft-MVCs might direct olfactory mucosal responses, we used an integrated single-cell transcriptional and protein analysis. Inhalation of Alternaria induced mucosal epithelial effector molecules including Chil4 and a distinct pathway leading to proliferation of the quiescent olfactory horizontal basal stem cell (HBC) pool, both triggered in the absence of olfactory apoptosis. Alternaria- and ATP-elicited HBC proliferation was dependent on TRPM5+ tuft-MVCs, identifying these specialized epithelial cells as regulators of olfactory stem cell responses. Together, our data provide high-resolution characterization of nasal tuft cell heterogeneity and identify a function of TRPM5+ tuft-MVCs in directing the olfactory mucosal response to allergens.
Collapse
Affiliation(s)
- Saltanat Ualiyeva
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Evan Lemire
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Caitlin Wong
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Alexander Perniss
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Amelia Boyd
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Evelyn C. Avilés
- Department of Neurobiology, Harvard Medical School, Boston, MA; currently at Faculty of Biological Sciences, Pontificia Universidad Católica de Chile
| | - Dante G. Minichetti
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Alice Maxfield
- Division of Otolaryngology-Head and Neck Surgery, Brigham and Women’s Hospital and Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA
| | - Rachel Roditi
- Division of Otolaryngology-Head and Neck Surgery, Brigham and Women’s Hospital and Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA
| | | | - Xin Wang
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Wenjiang Deng
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Nora A. Barrett
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Kathleen M. Buchheit
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Tanya M. Laidlaw
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Joshua A. Boyce
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Lora G. Bankova
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham & Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Adam L. Haber
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
17
|
Lyu R, Wu J, He Y, You Q, Qian Y, Jiang N, Cai Y, Chen D, Wu Z. Folate supports IL-25-induced tuft cell expansion following enteroviral infections. FASEB J 2024; 38:e23430. [PMID: 38243751 DOI: 10.1096/fj.202301928r] [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: 09/20/2023] [Revised: 12/11/2023] [Accepted: 01/03/2024] [Indexed: 01/21/2024]
Abstract
Intestinal tuft cells, a kind of epithelial immune cells, rapidly expand in response to pathogenic infections, which is associated with infection-induced interleukin 25 (IL-25) upregulation. However, the metabolic mechanism of IL-25-induced tuft cell expansion is largely unknown. Folate metabolism provides essential purine and methyl substrates for cell proliferation and differentiation. Thus, we aim to investigate the roles of folate metabolism playing in IL-25-induced tuft cell expansion by enteroviral infection and recombinant murine IL-25 (rmIL-25) protein-stimulated mouse models. At present, enteroviruses, such as EV71, CVA16, CVB3, and CVB4, upregulated IL-25 expression and induced tuft cell expansion in the intestinal tissues of mice. However, EV71 did not induce intestinal tuft cell expansion in IL-25-/- mice. Interestingly, compared to the mock group, folate was enriched in the intestinal tissues of both the EV71-infected group and the rmIL-25 protein-stimulated group. Moreover, folate metabolism supported IL-25-induced tuft cell expansion since both folate-depletion and anti-folate MTX-treated mice had a disrupted tuft cell expansion in response to rmIL-25 protein stimulation. In summary, our data suggested that folate metabolism supported intestinal tuft cell expansion in response to enterovirus-induced IL-25 expression, which provided a new insight into the mechanisms of tuft cell expansion from the perspective of folate metabolism.
Collapse
Affiliation(s)
- Ruining Lyu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Jing Wu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Yating He
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Qiao You
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Yajie Qian
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Na Jiang
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Yurong Cai
- School of Life Science, Ningxia University, Yinchuan, China
| | - Deyan Chen
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Zhiwei Wu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
- School of Life Science, Ningxia University, Yinchuan, China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
18
|
Li Y, Han M, Singh S, Breckenridge HA, Kreger JE, Stroupe CC, Sawicky DA, Kuo S, Goldsmith AM, Ke F, Shenoy AT, Bentley JK, Matsumoto I, Hershenson MB. Tuft cells are required for a rhinovirus-induced asthma phenotype in immature mice. JCI Insight 2024; 9:e166136. [PMID: 38061015 PMCID: PMC10906234 DOI: 10.1172/jci.insight.166136] [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: 10/10/2022] [Accepted: 12/05/2023] [Indexed: 01/17/2024] Open
Abstract
Infection of immature mice with rhinovirus (RV) induces an asthma-like phenotype consisting of type 2 inflammation, mucous metaplasia, eosinophilic inflammation, and airway hyperresponsiveness that is dependent on IL-25 and type 2 innate lymphoid cells (ILC2s). Doublecortin-like kinase 1-positive (DCLK1+) tuft cells are a major source of IL-25. We sought to determine the requirement of tuft cells for the RV-induced asthma phenotype in wild-type mice and mice deficient in Pou2f3, a transcription factor required for tuft cell development. C57BL/6J mice infected with RV-A1B on day 6 of life and RV-A2 on day 13 of life showed increased DCLK1+ tuft cells in the large airways. Compared with wild-type mice, RV-infected Pou2f3-/- mice showed reductions in IL-25 mRNA and protein expression, ILC2 expansion, type 2 cytokine expression, mucous metaplasia, lung eosinophils, and airway methacholine responsiveness. We conclude that airway tuft cells are required for the asthma phenotype observed in immature mice undergoing repeated RV infections. Furthermore, RV-induced tuft cell development provides a mechanism by which early-life viral infections could potentiate type 2 inflammatory responses to future infections.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Fang Ke
- Department of Microbiology and Immunology, and
| | - Anukul T. Shenoy
- Department of Microbiology and Immunology, and
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | - Marc B. Hershenson
- Department of Pediatrics
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
19
|
Purev E, Bahmed K, Kosmider B. Alveolar Organoids in Lung Disease Modeling. Biomolecules 2024; 14:115. [PMID: 38254715 PMCID: PMC10813493 DOI: 10.3390/biom14010115] [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: 07/26/2023] [Revised: 01/06/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Lung organoids display a tissue-specific functional phenomenon and mimic the features of the original organ. They can reflect the properties of the cells, such as morphology, polarity, proliferation rate, gene expression, and genomic profile. Alveolar type 2 (AT2) cells have a stem cell potential in the adult lung. They produce and secrete pulmonary surfactant and proliferate to restore the epithelium after damage. Therefore, AT2 cells are used to generate alveolar organoids and can recapitulate distal lung structures. Also, AT2 cells in human-induced pluripotent stem cell (iPSC)-derived alveolospheres express surfactant proteins and other factors, indicating their application as suitable models for studying cell-cell interactions. Recently, they have been utilized to define mechanisms of disease development, such as COVID-19, lung cancer, idiopathic pulmonary fibrosis, and chronic obstructive pulmonary disease. In this review, we show lung organoid applications in various pulmonary diseases, drug screening, and personalized medicine. In addition, stem cell-based therapeutics and approaches relevant to lung repair were highlighted. We also described the signaling pathways and epigenetic regulation of lung regeneration. It is critical to identify novel regulators of alveolar organoid generations to promote lung repair in pulmonary diseases.
Collapse
Affiliation(s)
- Enkhee Purev
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA
| | - Karim Bahmed
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
| | - Beata Kosmider
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
- Department of Cardiovascular Sciences, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
20
|
Kageyama T, Ito T, Tanaka S, Nakajima H. Physiological and immunological barriers in the lung. Semin Immunopathol 2024; 45:533-547. [PMID: 38451292 PMCID: PMC11136722 DOI: 10.1007/s00281-024-01003-y] [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: 10/04/2023] [Accepted: 02/10/2024] [Indexed: 03/08/2024]
Abstract
The lungs serve as the primary organ for respiration, facilitating the vital exchange of gases with the bloodstream. Given their perpetual exposure to external particulates and pathogens, they possess intricate protective barriers. Cellular adhesion in the lungs is robustly maintained through tight junctions, adherens junctions, and desmosomes. Furthermore, the pulmonary system features a mucociliary clearance mechanism that synthesizes mucus and transports it to the outside. This mucus is enriched with chemical barriers like antimicrobial proteins and immunoglobulin A (IgA). Additionally, a complex immunological network comprising epithelial cells, neural cells, and immune cells plays a pivotal role in pulmonary defense. A comprehensive understanding of these protective systems offers valuable insights into potential pathologies and their therapeutic interventions.
Collapse
Affiliation(s)
- Takahiro Kageyama
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba, 260-8670, Japan.
- Institute for Advanced Academic Research, Chiba University, Chiba, Japan.
| | - Takashi Ito
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba, 260-8670, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba, Japan
| | - Shigeru Tanaka
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba, 260-8670, Japan
| | - Hiroshi Nakajima
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba, 260-8670, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba, Japan
| |
Collapse
|
21
|
Graf J, Trautmann-Rodriguez M, Sabnis S, Kloxin AM, Fromen CA. On the path to predicting immune responses in the lung: Modeling the pulmonary innate immune system at the air-liquid interface (ALI). Eur J Pharm Sci 2023; 191:106596. [PMID: 37770004 PMCID: PMC10658361 DOI: 10.1016/j.ejps.2023.106596] [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: 06/12/2023] [Revised: 09/01/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023]
Abstract
Chronic respiratory diseases and infections are among the largest contributors to death globally, many of which still have no cure, including chronic obstructive pulmonary disorder, idiopathic pulmonary fibrosis, and respiratory syncytial virus among others. Pulmonary therapeutics afford untapped potential for treating lung infection and disease through direct delivery to the site of action. However, the ability to innovate new therapeutic paradigms for respiratory diseases will rely on modeling the human lung microenvironment and including key cellular interactions that drive disease. One key feature of the lung microenvironment is the air-liquid interface (ALI). ALI interface modeling techniques, using cell-culture inserts, organoids, microfluidics, and precision lung slices (PCLS), are rapidly developing; however, one major component of these models is lacking-innate immune cell populations. Macrophages, neutrophils, and dendritic cells, among others, represent key lung cell populations, acting as the first responders during lung infection or injury. Innate immune cells respond to and modulate stromal cells and bridge the gap between the innate and adaptive immune system, controlling the bodies response to foreign pathogens and debris. In this article, we review the current state of ALI culture systems with a focus on innate immune cells and suggest ways to build on current models to add complexity and relevant immune cell populations.
Collapse
Affiliation(s)
- Jodi Graf
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | | | - Simone Sabnis
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA
| | - April M Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA; Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA.
| | - Catherine A Fromen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
22
|
Ding Y, Zhao Z, Cai H, Zhou Y, Chen H, Bai Y, Liu Z, Liu S, Zhou W. Single-cell sequencing analysis related to sphingolipid metabolism guides immunotherapy and prognosis of skin cutaneous melanoma. Front Immunol 2023; 14:1304466. [PMID: 38077400 PMCID: PMC10701528 DOI: 10.3389/fimmu.2023.1304466] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 11/07/2023] [Indexed: 12/18/2023] Open
Abstract
Background We explore sphingolipid-related genes (SRGs) in skin melanoma (SKCM) to develop a prognostic indicator for patient outcomes. Dysregulated lipid metabolism is linked to aggressive behavior in various cancers, including SKCM. However, the exact role and mechanism of sphingolipid metabolism in melanoma remain partially understood. Methods We integrated scRNA-seq data from melanoma patients sourced from the GEO database. Through the utilization of the Seurat R package, we successfully identified distinct gene clusters associated with patient survival in the scRNA-seq data. Key prognostic genes were identified through single-factor Cox analysis and used to develop a prognostic model using LASSO and stepwise regression algorithms. Additionally, we evaluated the predictive potential of these genes within the immune microenvironment and their relevance to immunotherapy. Finally, we validated the functional significance of the high-risk gene IRX3 through in vitro experiments. Results Analysis of scRNA-seq data identified distinct expression patterns of 4 specific genes (SRGs) in diverse cell subpopulations. Re-clustering cells based on increased SRG expression revealed 7 subgroups with significant prognostic implications. Using marker genes, lasso, and Cox regression, we selected 11 genes to construct a risk signature. This signature demonstrated a strong correlation with immune cell infiltration and stromal scores, highlighting its relevance in the tumor microenvironment. Functional studies involving IRX3 knockdown in A375 and WM-115 cells showed significant reductions in cell viability, proliferation, and invasiveness. Conclusion SRG-based risk signature holds promise for precise melanoma prognosis. An in-depth exploration of SRG characteristics offers insights into immunotherapy response. Therapeutic targeting of the IRX3 gene may benefit melanoma patients.
Collapse
Affiliation(s)
- Yantao Ding
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Zhijie Zhao
- Department of Plastic Surgery, The Ninth Affiliated Hospital of Shanghai Jiaotong University, Shanghai, China
| | - Huabao Cai
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yi Zhou
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - He Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yun Bai
- Department of Plastic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhenran Liu
- Department of Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Shengxiu Liu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Wenming Zhou
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| |
Collapse
|
23
|
Ding L, Weygant N, Ding C, Lai Y, Li H. DCLK1 and tuft cells: Immune-related functions and implications for cancer immunotherapy. Crit Rev Oncol Hematol 2023; 191:104118. [PMID: 37660932 DOI: 10.1016/j.critrevonc.2023.104118] [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: 05/08/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/05/2023] Open
Abstract
DCLK1, a tuft cell marker, is widely expressed in various tumors. Its high expression levels are closely linked to malignant tumor progression, making it a potential tumor-related marker. Recent studies have shed light on the critical roles of DCLK1 and tuft cells in the immune response and the maintenance of epithelial homeostasis, as well as targeted immune escape mechanisms in the tumor microenvironment. This review aims to comprehensively examine the current understanding of immune-related functions mediated by DCLK1 and tuft cells in epithelial tissues, including the roles of relevant cells and important factors involved. Additionally, this review will discuss recent advances in anti-tumor immunity mediated by DCLK1/tuft cells and their potential as immunotherapeutic targets. Furthermore, we will consider the potential impact of DCLK1 targeted therapy in cancer immunotherapy, particularly DCLK1 kinase inhibitors as potential therapeutic drugs in anti-tumor immunity, providing a new perspective and reference for future research.
Collapse
Affiliation(s)
- Ling Ding
- Traditional Chinese Medicine Department, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Nathaniel Weygant
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Chenhuan Ding
- Traditional Chinese Medicine Department, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yi Lai
- Department of Head and Neck Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - He Li
- Traditional Chinese Medicine Department, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| |
Collapse
|
24
|
Chang C, Chen G, Wu W, Chen D, Chen S, Gao J, Feng Y, Zhen G. Exogenous IL-25 ameliorates airway neutrophilia via suppressing macrophage M1 polarization and the expression of IL-12 and IL-23 in asthma. Respir Res 2023; 24:260. [PMID: 37898756 PMCID: PMC10613395 DOI: 10.1186/s12931-023-02557-5] [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: 08/07/2023] [Accepted: 10/07/2023] [Indexed: 10/30/2023] Open
Abstract
BACKGROUND Severe asthma is associated with substantial mortality and has unmet therapeutic need. A subset of severe asthma is characterized by neutrophilic airway inflammation. Classically activated (or M1) macrophages which express IL-12 and IL-23 are associated with airway neutrophilia in asthma. Exogenous IL-25 was reported to suppress intestinal inflammation in animal models of inflammatory bowel diseases via suppressing IL-12 and IL-23 production. We hypothesize that IL-25 ameliorates airway neutrophilia via inhibiting macrophage M1 polarization and the expression of IL-12 and IL-23 in asthma. METHODS In a mouse model of neutrophil-dominant allergic airway inflammation, the effect of mouse recombinant IL-25 on airway inflammation were assessed by H&E staining and bronchoalveolar lavage (BAL) cell counting. The percentage of M1 macrophages in lung tissue and BAL cells were analyzed by flow cytometry. Quantitative PCR and immunostaining were performed to measure the expression of Il12, Il23, and inflammatory cytokines. Mechanistic experiments were performed in primary culture of macrophages from mouse lungs. The expression of IL-12, IL-23 and IL-25 in sputum was analyzed in a cohort of severe asthma and subjects with eosinophilic or non-eosinophilic asthma. RESULTS Intranasal administration of IL-25 markedly decreased the number of neutrophils in BAL cells in a murine model of neutrophil-dominant allergic airway inflammation. Moreover, exogenous IL-25 decreased the number of M1 macrophages, and reduced the expression of IL-12, IL-23 in the lungs of the mouse model. Exogenous IL-25 also inhibited the expression of inflammatory cytokines IL-1β, IFN-γ, TNF-α and IL-17 A. In vitro, IL-25 suppressed IL-12 and IL-23 expression in lipopolysaccharide (LPS)-stimulated primary culture of mouse pulmonary macrophages. Mechanistically, IL-25 inhibited LPS-induced c-Rel translocation to nucleus via STAT3-dependent signaling. In a cohort of severe asthma, IL-25 protein levels in sputum were significantly lower than control subjects. The transcript levels of IL-12 and IL-23 were increased whereas IL-25 transcripts were decreased in sputum cells from subjects with non-eosinophilic asthma compared to eosinophilic asthma. CONCLUSIONS IL-25 expression is downregulated in subjects with severe or non-eosinophilic asthma. Exogenous IL-25 ameliorates airway neutrophilia, at least in part, via inhibiting macrophage M1 polarization and the expression of IL-12 and IL-23.
Collapse
Affiliation(s)
- Chenli Chang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China
| | - Gongqi Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China
| | - Wenliang Wu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China
| | - Dian Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China
| | - Shengchong Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China
| | - Jiali Gao
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China
| | - Yuchen Feng
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China.
- Division of Pulmonary and Critical Care Medicine, Tongji Hospital, 1095 Jiefang Avenue, 430030, Wuhan, China.
| | - Guohua Zhen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China.
- Division of Respiratory and Critical Care Medicine, Tongji Hospital, 430030, Wuhan, China.
| |
Collapse
|
25
|
Ortiz-Carpena JF, Inclan-Rico JM, Pastore CF, Hung LY, Wilkerson WB, Weiner MB, Lin C, Gentile ME, Cohen NA, Saboor IA, Vaughan AE, Rossi HL, Herbert DR. [WITHDRAWN] Neuron-dependent tuft cell expansion initiates sinonasal allergic Type 2 inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.04.547596. [PMID: 37461610 PMCID: PMC10349937 DOI: 10.1101/2023.07.04.547596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/24/2023]
Abstract
The authors have withdrawn this manuscript owing to inaccuracies in the calculation of tuft cell numbers and errors in the selection of immunofluorescence images used to support our claims. Therefore, the authors do not wish this work to be cited as reference for the project. If you have any questions, please contact the corresponding author.
Collapse
|
26
|
Orieux A, Enaud R, Imbert S, Boyer P, Begot E, Camino A, Boyer A, Berger P, Gruson D, Delhaes L, Prevel R. The gut microbiota composition is linked to subsequent occurrence of ventilator-associated pneumonia in critically ill patients. Microbiol Spectr 2023; 11:e0064123. [PMID: 37713505 PMCID: PMC10581192 DOI: 10.1128/spectrum.00641-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 07/26/2023] [Indexed: 09/17/2023] Open
Abstract
Ventilator-associated pneumonia (VAP) is the most frequent nosocomial infection in critically ill-ventilated patients. Oropharyngeal and lung microbiota have been demonstrated to be associated with VAP occurrence, but the involvement of gut microbiota has not been investigated so far. Therefore, the aim of this study is to compare the composition of the gut microbiota between patients who subsequently develop VAP and those who do not. A rectal swab was performed at admission of every consecutive patient into the intensive care unit (ICU) from October 2019 to March 2020. After DNA extraction, V3-V4 and internal transcribed spacer 2 regions deep-sequencing was performed on MiSeq sequencer (Illumina) and data were analyzed using Divisive Amplicon Denoising Algorithm 2 (DADA2) pipeline. Among 255 patients screened, 42 (16%) patients with invasive mechanical ventilation for more than 48 h were included, 18 (43%) with definite VAP and 24 without (57%). Patients who later developed VAP had similar gut bacteriobiota and mycobiota α-diversities compared to those who did not develop VAP. However, gut mycobiota was dissimilar (β-diversity) between these two groups. The presence of Megasphaera massiliensis was associated with the absence of VAP occurrence, whereas the presence of the fungal genus Alternaria sp. was associated with the occurrence of VAP. The composition of the gut microbiota, but not α-diversity, differs between critically ill patients who subsequently develop VAP and those who do not. This study encourages large multicenter cohort studies investigating the role of gut-lung axis and oropharyngeal colonization in the development of VAP in ICU patients. Trial registration number: NCT04131569, date of registration: 18 October 2019. IMPORTANCE The composition of the gut microbiota, but not α-diversity, differs between critically ill patients who subsequently develop ventilator-associated pneumonia (VAP) and those who do not. Investigating gut microbiota composition could help to tailor probiotics to provide protection against VAP.
Collapse
Affiliation(s)
- Arthur Orieux
- CHU Bordeaux, Medical Intensive Care Unit, Bordeaux, France
| | - Raphaël Enaud
- Univ Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Inserm UMR 1045, Bordeaux, France
- CHU Bordeaux, CRCM Pédiatrique, Bordeaux, France
| | - Sébastien Imbert
- Univ Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Inserm UMR 1045, Bordeaux, France
- Mycology-Parasitology Department, CHU Bordeaux, Bordeaux, France
| | - Philippe Boyer
- CHU Bordeaux, Medical Intensive Care Unit, Bordeaux, France
| | - Erwan Begot
- CHU Bordeaux, Medical Intensive Care Unit, Bordeaux, France
| | - Adrian Camino
- Univ Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Inserm UMR 1045, Bordeaux, France
| | - Alexandre Boyer
- CHU Bordeaux, Medical Intensive Care Unit, Bordeaux, France
- Univ Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Inserm UMR 1045, Bordeaux, France
| | - Patrick Berger
- Univ Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Inserm UMR 1045, Bordeaux, France
| | - Didier Gruson
- CHU Bordeaux, Medical Intensive Care Unit, Bordeaux, France
- Univ Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Inserm UMR 1045, Bordeaux, France
| | - Laurence Delhaes
- Univ Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Inserm UMR 1045, Bordeaux, France
- Mycology-Parasitology Department, CHU Bordeaux, Bordeaux, France
| | - Renaud Prevel
- CHU Bordeaux, Medical Intensive Care Unit, Bordeaux, France
- Univ Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Inserm UMR 1045, Bordeaux, France
| |
Collapse
|
27
|
Abstract
The worldwide prevalence of asthma and allergic disorders (allergic rhinitis, atopic dermatitis, food allergy) has been steadily rising in recent decades. It is now estimated that up to 20% of the global population is afflicted by an allergic disease, with increasing incidence rates in both high- and low-income countries. The World Allergy Organization estimates that the total economic burden of asthma and allergic rhinitis alone is approximately $21 billion per year. While allergic stimuli are a complex and heterogenous class of inputs including parasites, pollens, food antigens, drugs, and metals, it has become clear that fungi are major drivers of allergic disease, with estimates that fungal sensitization occurs in 20-30% of atopic individuals and up to 80% of asthma patients. Fungi are eukaryotic microorganisms that can be found throughout the world in high abundance in both indoor and outdoor environments. Understanding how and why fungi act as triggers of allergic type 2 inflammation will be crucial for combating this important health problem. In recent years, there have been significant advances in our understanding of fungi-induced type 2 immunity, however there is still much we don't understand, including why fungi have a tendency to induce allergic reactions in the first place. Here, we will discuss how fungi trigger type 2 immune responses and posit why this response has been evolutionarily selected for induction during fungal encounter.
Collapse
Affiliation(s)
- Yufan Zheng
- Molecular Mycology and Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Eric V. Dang
- Molecular Mycology and Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
28
|
Dos Santos HT, Nam K, Small T, Maslow FM, Baker OJ. Confirming the Identity of Tuft Cells in Mouse Submandibular Glands. GASTRO HEP ADVANCES 2023; 2:1053-1055. [PMID: 38098741 PMCID: PMC10720652 DOI: 10.1016/j.gastha.2023.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Affiliation(s)
- H Tavares Dos Santos
- Department of Otolaryngology-Head and Neck Surgery, University of Missouri, Columbia, Missouri
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
| | - K Nam
- Department of Otolaryngology-Head and Neck Surgery, University of Missouri, Columbia, Missouri
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
| | - T Small
- Department of Otolaryngology-Head and Neck Surgery, University of Missouri, Columbia, Missouri
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
| | - F M Maslow
- Department of Otolaryngology-Head and Neck Surgery, University of Missouri, Columbia, Missouri
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
| | - O J Baker
- Department of Otolaryngology-Head and Neck Surgery, University of Missouri, Columbia, Missouri
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
- Department of Biochemistry, University of Missouri, Columbia, Missouri
| |
Collapse
|
29
|
Kühl L, Graichen P, von Daacke N, Mende A, Wygrecka M, Potaczek DP, Miethe S, Garn H. Human Lung Organoids-A Novel Experimental and Precision Medicine Approach. Cells 2023; 12:2067. [PMID: 37626876 PMCID: PMC10453737 DOI: 10.3390/cells12162067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/31/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
The global burden of respiratory diseases is very high and still on the rise, prompting the need for accurate models for basic and translational research. Several model systems are currently available ranging from simple airway cell cultures to complex tissue-engineered lungs. In recent years, human lung organoids have been established as highly transferrable three-dimensional in vitro model systems for lung research. For acute infectious and chronic inflammatory diseases as well as lung cancer, human lung organoids have opened possibilities for precise in vitro research and a deeper understanding of mechanisms underlying lung injury and regeneration. Human lung organoids from induced pluripotent stem cells or from adult stem cells of patients' samples introduce tools for understanding developmental processes and personalized medicine approaches. When further state-of-the-art technologies and protocols come into use, the full potential of human lung organoids can be harnessed. High-throughput assays in drug development, gene therapy, and organoid transplantation are current applications of organoids in translational research. In this review, we emphasize novel approaches in translational and personalized medicine in lung research focusing on the use of human lung organoids.
Collapse
Affiliation(s)
- Laura Kühl
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University of Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (L.K.); (P.G.); (N.v.D.); (A.M.); (D.P.P.)
| | - Pauline Graichen
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University of Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (L.K.); (P.G.); (N.v.D.); (A.M.); (D.P.P.)
| | - Nele von Daacke
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University of Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (L.K.); (P.G.); (N.v.D.); (A.M.); (D.P.P.)
| | - Anne Mende
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University of Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (L.K.); (P.G.); (N.v.D.); (A.M.); (D.P.P.)
| | - Malgorzata Wygrecka
- Center for Infection and Genomics of the Lung (CIGL), Universities of Giessen and Marburg Lung Center (UGMLC), 35392 Giessen, Germany;
- Institute of Lung Health, German Center for Lung Research (DZL), 35392 Giessen, Germany
- CSL Behring Innovation GmbH, 35041 Marburg, Germany
| | - Daniel P. Potaczek
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University of Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (L.K.); (P.G.); (N.v.D.); (A.M.); (D.P.P.)
- Center for Infection and Genomics of the Lung (CIGL), Universities of Giessen and Marburg Lung Center (UGMLC), 35392 Giessen, Germany;
- Bioscientia MVZ Labor Mittelhessen GmbH, 35394 Giessen, Germany
| | - Sarah Miethe
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University of Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (L.K.); (P.G.); (N.v.D.); (A.M.); (D.P.P.)
| | - Holger Garn
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University of Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (L.K.); (P.G.); (N.v.D.); (A.M.); (D.P.P.)
| |
Collapse
|
30
|
Sun J, Li MX, Xie YM, Zhang YR, Chai YR. Thymic tuft cells: potential "regulators" of non-mucosal tissue development and immune response. Immunol Res 2023; 71:554-564. [PMID: 36961668 PMCID: PMC10037390 DOI: 10.1007/s12026-023-09372-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 03/09/2023] [Indexed: 03/25/2023]
Abstract
As the leading central immune organ, the thymus is where T cells differentiate and mature, and plays an essential regulatory role in the adaptive immune response. Tuft cells, as chemosensory cells, were first found in rat tracheal epithelial, later gradually confirmed to exist in various mucosal and non-mucosal tissues. Although tuft cells are epithelial-derived, because of their wide heterogeneity, they show functions similar to cholinergic and immune cells in addition to chemosensory ability. As newly discovered non-mucosal tuft cells, thymic tuft cells have been demonstrated to be involved in and play vital roles in immune responses such as antigen presentation, immune tolerance, and type 2 immunity. In addition to their unique functions in the thymus, thymic tuft cells have the characteristics of peripheral tuft cells, so they may also participate in the process of tumorigenesis and virus infection. Here, we review tuft cells' characteristics, distribution, and potential functions. More importantly, the potential role of thymic tuft cells in immune response, tumorigenesis, and severe acute respiratory syndrome coronavirus 2(SARS-CoV-2) infection was summarized and discussed.
Collapse
Affiliation(s)
- Jun Sun
- Medical School of Zhengzhou University, Zhengzhou, Henan Province, 450052, People's Republic of China
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, People's Republic of China
| | - Ming-Xin Li
- Medical School of Zhengzhou University, Zhengzhou, Henan Province, 450052, People's Republic of China
| | - Yi-Meng Xie
- School of Fine Arts of Zhengzhou University, Zhengzhou, Henan Province, 450001, People's Republic of China
| | - Ya-Ru Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, No.100, Kexuedadao Road, Zhengzhou, Henan Province, 450001, People's Republic of China
| | - Yu-Rong Chai
- Department of Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, No.100, Kexuedadao Road, Zhengzhou, Henan Province, 450001, People's Republic of China.
| |
Collapse
|
31
|
Kotas ME, Patel NN, Cope EK, Gurrola JG, Goldberg AN, Pletcher SD, Seibold MA, Moore CM, Gordon ED. IL-13-associated epithelial remodeling correlates with clinical severity in nasal polyposis. J Allergy Clin Immunol 2023; 151:1277-1285. [PMID: 36736797 PMCID: PMC10243183 DOI: 10.1016/j.jaci.2022.12.826] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/19/2022] [Accepted: 12/28/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Epithelial remodeling is a histopathologic feature of chronic inflammatory airway diseases including chronic rhinosinusitis (CRS). Cell-type shifts and their relationship to CRS endotypes and severity are incompletely described. OBJECTIVE We sought to understand the relationship of epithelial cell remodeling to inflammatory endotypes and disease outcomes in CRS. METHODS Using cell-type transcriptional signatures derived from epithelial single-cell sequencing, we analyzed bulk RNA-sequencing data from sinus epithelial brushings obtained from patients with CRS with and without nasal polyps in comparison to healthy controls. RESULTS The airway epithelium in nasal polyposis displayed increased tuft cell transcripts and decreased ciliated cell transcripts along with an IL-13 activation signature. In contrast, CRS without polyps showed an IL-17 activation signature. IL-13 activation scores were associated with increased tuft cell, goblet cell, and mast cell scores and decreased ciliated cell scores. Furthermore, the IL-13 score was strongly associated with a previously reported activated ("polyp") tuft cell score and a prostaglandin E2 activation signature. The Lund-Mackay score, a computed tomographic metric of sinus opacification, correlated positively with activated tuft cell, mast cell, prostaglandin E2, and IL-13 signatures and negatively with ciliated cell transcriptional signatures. CONCLUSIONS These results demonstrate that cell-type alterations and prostaglandin E2 stimulation are key components of IL-13-induced epithelial remodeling in nasal polyposis, whereas IL-17 signaling is more prominent in CRS without polyps, and that clinical severity correlates with the degree of IL-13-driven epithelial remodeling.
Collapse
Affiliation(s)
- Maya E Kotas
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, Calif
| | - Neil N Patel
- Department of Otolaryngology - Head and Neck Surgery, University of California, San Francisco, Calif
| | - Emily K Cope
- Center for Applied Microbiome Sciences, the Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Ariz
| | - Jose G Gurrola
- Department of Otolaryngology - Head and Neck Surgery, University of California, San Francisco, Calif
| | - Andrew N Goldberg
- Department of Otolaryngology - Head and Neck Surgery, University of California, San Francisco, Calif
| | - Steven D Pletcher
- Department of Otolaryngology - Head and Neck Surgery, University of California, San Francisco, Calif; Surgical Service, ENT Section, San Francisco VA Medical Center, San Francisco, Calif
| | - Max A Seibold
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colo; Department of Pediatrics, National Jewish Health, Denver, Colo; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colo
| | - Camille M Moore
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colo; Department of Biostatistics and Informatics, University of Colorado, Aurora, Colo.
| | - Erin D Gordon
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, Calif.
| |
Collapse
|
32
|
Schroeder JH, Beattie G, Lo JW, Zabinski T, Powell N, Neves JF, Jenner RG, Lord GM. CD90 is not constitutively expressed in functional innate lymphoid cells. Front Immunol 2023; 14:1113735. [PMID: 37114052 PMCID: PMC10126679 DOI: 10.3389/fimmu.2023.1113735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/28/2023] [Indexed: 04/29/2023] Open
Abstract
Huge progress has been made in understanding the biology of innate lymphoid cells (ILC) by adopting several well-known concepts in T cell biology. As such, flow cytometry gating strategies and markers, such as CD90, have been applied to indentify ILC. Here, we report that most non-NK intestinal ILC have a high expression of CD90 as expected, but surprisingly a sub-population of cells exhibit low or even no expression of this marker. CD90-negative and CD90-low CD127+ ILC were present amongst all ILC subsets in the gut. The frequency of CD90-negative and CD90-low CD127+ ILC was dependent on stimulatory cues in vitro and enhanced by dysbiosis in vivo. CD90-negative and CD90-low CD127+ ILC were a potential source of IL-13, IFNγ and IL-17A at steady state and upon dysbiosis- and dextran sulphate sodium-elicited colitis. Hence, this study reveals that, contrary to expectations, CD90 is not constitutively expressed by functional ILC in the gut.
Collapse
Affiliation(s)
- Jan-Hendrik Schroeder
- School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Gordon Beattie
- Cancer Research UK (CRUK) City of London Centre Single Cell Genomics Facility, University College London Cancer Institute, University College London (UCL), London, United Kingdom
- Genomics Translational Technology Platform, University College London (UCL) Cancer Institute, University College London, London, United Kingdom
| | - Jonathan W. Lo
- School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Tomasz Zabinski
- School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Nick Powell
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Joana F. Neves
- School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Richard G. Jenner
- University College London (UCL) Cancer Institute, University College London, London, United Kingdom
| | - Graham M. Lord
- School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
33
|
Laidlaw TM, Boyce JA. Updates on immune mechanisms in aspirin-exacerbated respiratory disease. J Allergy Clin Immunol 2023; 151:301-309. [PMID: 36184313 PMCID: PMC9905222 DOI: 10.1016/j.jaci.2022.08.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/03/2022] [Accepted: 08/10/2022] [Indexed: 02/07/2023]
Abstract
Aspirin-exacerbated respiratory disease has fascinated and frustrated specialists in allergy/immunology, pulmonology, and otorhinolaryngology for decades. It generally develops in previously healthy young adults and is unremitting and challenging to treat. The classical triad of asthma, nasal polyposis, and pathognomonic respiratory reactions to aspirin and other cyclooxygenase-1 inhibitors is accompanied by high levels of mast cell activation, cysteinyl leukotriene production, platelet activation, and severe type 2 respiratory inflammation. The "unbraking" of mast cell activation and further cysteinyl leukotriene generation induced by cyclooxygenase-1 inhibition reflect an idiosyncratic dependency on cyclooxygenase-1-derived products, likely prostaglandin E2, to maintain a tenuous homeostasis. Although cysteinyl leukotrienes are clear disease effectors, little else was known about their cellular sources and targets, and the contributions from other mediators and type 2 respiratory inflammation effector cells to disease pathophysiology were unknown until recently. The applications of targeted biological therapies, single-cell genomics, and transgenic animal approaches have substantially advanced our understanding of aspirin-exacerbated respiratory disease pathogenesis and treatment and have also revealed disease heterogeneity. This review covers novel insights into the immunopathogenesis of aspirin-exacerbated respiratory disease from each of these lines of research, including the roles of lipid mediators, effector cell populations, and inflammatory cytokines, discusses unanswered questions regarding cause and pathogenesis, and considers potential future therapeutic options.
Collapse
Affiliation(s)
- Tanya M Laidlaw
- Department of Medicine, the Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Harvard Medical School, Jeff and Penny Vinik Center for Translational Immunology Research, Boston, Mass.
| | - Joshua A Boyce
- Department of Medicine, the Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Harvard Medical School, Jeff and Penny Vinik Center for Translational Immunology Research, Boston, Mass
| |
Collapse
|
34
|
McDaniel MM, Lara HI, von Moltke J. Initiation of type 2 immunity at barrier surfaces. Mucosal Immunol 2023; 16:86-97. [PMID: 36642383 DOI: 10.1016/j.mucimm.2022.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 01/15/2023]
Abstract
Although seemingly unrelated, parasitic worms, venoms, and allergens all induce a type 2 immune response. The effector functions and clinical features of type 2 immunity are well-defined, but fundamental questions about the initiation of type 2 immunity remain unresolved. How are these enormously diverse type 2 stimuli first detected? How are type 2 helper T cells primed and regulated? And how do mechanisms of type 2 initiation vary across tissues? Here, we review the common themes governing type 2 immune sensing and explore aspects of T cell priming and effector reactivation that make type 2 helper T cells a unique T helper lineage. Throughout the review, we emphasize the importance of non-hematopoietic cells and highlight how the unique anatomy and physiology of each barrier tissue shape mechanisms of type 2 immune initiation.
Collapse
Affiliation(s)
- Margaret M McDaniel
- Department of Immunology, University of Washington School of Medicine, Seattle, USA.
| | - Heber I Lara
- Department of Immunology, University of Washington School of Medicine, Seattle, USA
| | - Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, Seattle, USA
| |
Collapse
|
35
|
Jiang W, Wang Z, Zhang J, Li M. Interleukin 25 and its biological features and function in intestinal diseases. Cent Eur J Immunol 2023; 47:362-372. [PMID: 36817397 PMCID: PMC9901255 DOI: 10.5114/ceji.2022.124416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 01/13/2023] [Indexed: 02/04/2023] Open
Abstract
Interleukin 25 (IL-25), also known as IL-17E, is a member of the IL-17 cytokine family and an important regulator of the type 2 immune response. Accumulating evidence suggests that IL-25 interacts with diverse immune as well as non-immune cells and plays a rather complicated role in different backgrounds of multiple organs. IL-25 has been studied in the physiology and pathology of the intestine to some extent. With epithelial cells being an important source in the intestine, IL-25 plays a key role in intestinal immune responses and is associated with inappropriate allergic reactions, autoimmune diseases, and cancer tumorigenesis. In this review, we discuss the emerging comprehension of the biology of IL-25, as well as its cellular sources, targets, and signaling transduction. In particular, we discuss how IL-25 participates in the development of intestinal diseases including helminth infection, inflammatory bowel diseases, food allergy and colorectal cancer, as well as its underlying role in future therapy.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zehui Wang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jun Zhang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Minghui Li
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
36
|
Kotas ME, O'Leary CE, Locksley RM. Tuft Cells: Context- and Tissue-Specific Programming for a Conserved Cell Lineage. ANNUAL REVIEW OF PATHOLOGY 2023; 18:311-335. [PMID: 36351364 PMCID: PMC10443898 DOI: 10.1146/annurev-pathol-042320-112212] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Tuft cells are found in tissues with distinct stem cell compartments, tissue architecture, and luminal exposures but converge on a shared transcriptional program, including expression of taste transduction signaling pathways. Here, we summarize seminal and recent findings on tuft cells, focusing on major categories of function-instigation of type 2 cytokine responses, orchestration of antimicrobial responses, and emerging roles in tissue repair-and describe tuft cell-derived molecules used to affect these functional programs. We review what is known about the development of tuft cells from epithelial progenitors under homeostatic conditions and during disease. Finally, we discuss evidence that immature, or nascent, tuft cells with potential for diverse functions are driven toward dominant effector programs by tissue- or perturbation-specific contextual cues, which may result in heterogeneous mature tuft cell phenotypes both within and between tissues.
Collapse
Affiliation(s)
- Maya E Kotas
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, California, USA
- Department of Medicine, University of California, San Francisco, California, USA
| | - Claire E O'Leary
- Department of Medicine, University of California, San Francisco, California, USA
- Current affiliation: Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Richard M Locksley
- Department of Medicine, University of California, San Francisco, California, USA
- Department of Microbiology and Immunology, University of California, San Francisco, California, USA;
- Howard Hughes Medical Institute, University of California, San Francisco, California, USA
| |
Collapse
|
37
|
Li L, Ma M, Duan T, Sui X. The critical roles and therapeutic implications of tuft cells in cancer. Front Pharmacol 2022; 13:1047188. [PMID: 36569325 PMCID: PMC9780677 DOI: 10.3389/fphar.2022.1047188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Tuft cells are solitary chemosensory epithelial cells with microvilli at the top, which are found in hollow organs such as the gastrointestinal tract, pancreas, and lungs. Recently, an increasing number of studies have revealed the chemotactic abilities and immune function of the tuft cells, and numerous efforts have been devoted to uncovering the role of tuft cells in tumors. Notably, accumulating evidence has shown that the specific genes (POU2F3, DCLK1) expressed in tuft cells are involved in vital processes related with carcinogenesis and cancer development. However, the interaction between the tuft cells and cancer remains to be further elucidated. Here, based on an introduction of biological functions and specific markers of the tuft cells, we have summarized the functional roles and potential therapeutic implications of tuft cells in cancers, including pancreatic cancer, lung cancer, gastric cancer, colon cancer, and liver cancer, which is in the hope of inspiring the future research in validating tuft cells as novel strategies for cancer therapies.
Collapse
Affiliation(s)
- Lin Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Mengmeng Ma
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Ting Duan
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Xinbing Sui
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
- Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
38
|
Varricchi G, Ferri S, Pepys J, Poto R, Spadaro G, Nappi E, Paoletti G, Virchow JC, Heffler E, Canonica WG. Biologics and airway remodeling in severe asthma. Allergy 2022; 77:3538-3552. [PMID: 35950646 PMCID: PMC10087445 DOI: 10.1111/all.15473] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/03/2022] [Accepted: 08/05/2022] [Indexed: 01/28/2023]
Abstract
Asthma is a chronic inflammatory airway disease resulting in airflow obstruction, which in part can become irreversible to conventional therapies, defining the concept of airway remodeling. The introduction of biologics in severe asthma has led in some patients to the complete normalization of previously considered irreversible airflow obstruction. This highlights the need to distinguish a "fixed" airflow obstruction due to structural changes unresponsive to current therapies, from a "reversible" one as demonstrated by lung function normalization during biological therapies not previously obtained even with high-dose systemic glucocorticoids. The mechanisms by which exposure to environmental factors initiates the inflammatory responses that trigger airway remodeling are still incompletely understood. Alarmins represent epithelial-derived cytokines that initiate immunologic events leading to inflammatory airway remodeling. Biological therapies can improve airflow obstruction by addressing these airway inflammatory changes. In addition, biologics might prevent and possibly even revert "fixed" remodeling due to structural changes. Hence, it appears clinically important to separate the therapeutic effects (early and late) of biologics as a new paradigm to evaluate the effects of these drugs and future treatments on airway remodeling in severe asthma.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization (WAO) Center of Excellence, Naples, Italy.,Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, Naples, Italy
| | - Sebastian Ferri
- Personalized Medicine Asthma and Allergy Unit - IRCCS Humanitas Research Hospital, Milan, Italy
| | - Jack Pepys
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization (WAO) Center of Excellence, Naples, Italy
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization (WAO) Center of Excellence, Naples, Italy
| | - Emanuele Nappi
- Personalized Medicine Asthma and Allergy Unit - IRCCS Humanitas Research Hospital, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Giovanni Paoletti
- Personalized Medicine Asthma and Allergy Unit - IRCCS Humanitas Research Hospital, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | | | - Enrico Heffler
- Personalized Medicine Asthma and Allergy Unit - IRCCS Humanitas Research Hospital, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Walter G Canonica
- Personalized Medicine Asthma and Allergy Unit - IRCCS Humanitas Research Hospital, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
39
|
Duchesne M, Okoye I, Lacy P. Epithelial cell alarmin cytokines: Frontline mediators of the asthma inflammatory response. Front Immunol 2022; 13:975914. [PMID: 36311787 PMCID: PMC9616080 DOI: 10.3389/fimmu.2022.975914] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/20/2022] [Indexed: 12/02/2022] Open
Abstract
The exposure of the airway epithelium to external stimuli such as allergens, microbes, and air pollution triggers the release of the alarmin cytokines IL-25, IL-33 and thymic stromal lymphopoietin (TSLP). IL-25, IL-33 and TSLP interact with their ligands, IL-17RA, IL1RL1 and TSLPR respectively, expressed by hematopoietic and non-hematopoietic cells including dendritic cells, ILC2 cells, endothelial cells, and fibroblasts. Alarmins play key roles in driving type 2-high, and to a lesser extent type 2-low responses, in asthma. In addition, studies in which each of these three alarmins were targeted in allergen-challenged mice showed decreased chronicity of type-2 driven disease. Consequently, ascertaining the mechanism of activity of these upstream mediators has implications for understanding the outcome of targeted therapies designed to counteract their activity and alleviate downstream type 2-high and low effector responses. Furthermore, identifying the factors which shift the balance between the elicitation of type 2-high, eosinophilic asthma and type-2 low, neutrophilic-positive/negative asthma by alarmins is essential. In support of these efforts, observations from the NAVIGATOR trial imply that targeting TSLP in patients with tezepelumab results in reduced asthma exacerbations, improved lung function and control of the disease. In this review, we will discuss the mechanisms surrounding the secretion of IL-25, IL-33, and TSLP from the airway epithelium and how this influences the allergic airway cascade. We also review in detail how alarmin-receptor/co-receptor interactions modulate downstream allergic inflammation. Current strategies which target alarmins, their efficacy and inflammatory phenotype will be discussed.
Collapse
|
40
|
Tavares dos Santos H, Nam K, Maslow FM, Small T, Galloway TL, Dooley LM, Tassone PT, Zitsch RP, Weisman GA, Baker OJ. Tuft Cells Are Present in Submandibular Glands Across Species. J Histochem Cytochem 2022; 70:659-667. [PMID: 35993302 PMCID: PMC9527474 DOI: 10.1369/00221554221120301] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/27/2022] [Indexed: 11/22/2022] Open
Abstract
Tuft cells are bottle-shaped, microvilli-projecting chemosensory cells located in the lining of a variety of epithelial tissues and, following their identification approximately 60 years ago, have been linked to immune system function in a variety of epithelia. Until recently, Tuft cells had not been convincingly demonstrated to be present in salivary glands with their detection by transmission electron microscopy only shown in a handful of earlier studies using rat salivary glands, and no follow-up work has been conducted to verify their presence in salivary glands of other species. Here, we demonstrate that Tuft cells are present in the submandibular glands of various species (i.e., mouse, pig and human) using transmission electron microscopy and confocal immunofluorescent analysis for the POU class 2 homeobox 3 (POU2F3), which is considered to be a master regulator of Tuft cell identity.
Collapse
Affiliation(s)
- Harim Tavares dos Santos
- Department of Otolaryngology-Head and Neck
Surgery, University of Missouri, Columbia, Missouri
- Christopher S. Bond Life Sciences Center,
University of Missouri, Columbia, Missouri
| | - Kihoon Nam
- Department of Otolaryngology-Head and Neck
Surgery, University of Missouri, Columbia, Missouri
- Christopher S. Bond Life Sciences Center,
University of Missouri, Columbia, Missouri
| | - Frank M. Maslow
- Department of Otolaryngology-Head and Neck
Surgery, University of Missouri, Columbia, Missouri
- Christopher S. Bond Life Sciences Center,
University of Missouri, Columbia, Missouri
| | - Travis Small
- Department of Otolaryngology-Head and Neck
Surgery, University of Missouri, Columbia, Missouri
- Christopher S. Bond Life Sciences Center,
University of Missouri, Columbia, Missouri
| | - Tabitha L.I. Galloway
- Department of Otolaryngology-Head and Neck
Surgery, University of Missouri, Columbia, Missouri
| | - Laura M. Dooley
- Department of Otolaryngology-Head and Neck
Surgery, University of Missouri, Columbia, Missouri
| | - Patrick T. Tassone
- Department of Otolaryngology-Head and Neck
Surgery, University of Missouri, Columbia, Missouri
| | - Robert P. Zitsch
- Department of Otolaryngology-Head and Neck
Surgery, University of Missouri, Columbia, Missouri
| | - Gary A. Weisman
- Department of Biochemistry, University of
Missouri, Columbia, Missouri
- Christopher S. Bond Life Sciences Center,
University of Missouri, Columbia, Missouri
| | - Olga J. Baker
- Department of Otolaryngology-Head and Neck
Surgery, University of Missouri, Columbia, Missouri
- Department of Biochemistry, University of
Missouri, Columbia, Missouri
- Christopher S. Bond Life Sciences Center,
University of Missouri, Columbia, Missouri
| |
Collapse
|
41
|
Kummerlowe C, Mwakamui S, Hughes TK, Mulugeta N, Mudenda V, Besa E, Zyambo K, Shay JES, Fleming I, Vukovic M, Doran BA, Aicher TP, Wadsworth MH, Bramante JT, Uchida AM, Fardoos R, Asowata OE, Herbert N, Yilmaz ÖH, Kløverpris HN, Garber JJ, Ordovas-Montanes J, Gartner Z, Wallach T, Shalek AK, Kelly P. Single-cell profiling of environmental enteropathy reveals signatures of epithelial remodeling and immune activation. Sci Transl Med 2022; 14:eabi8633. [PMID: 36044598 PMCID: PMC9594855 DOI: 10.1126/scitranslmed.abi8633] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Environmental enteropathy (EE) is a subclinical condition of the small intestine that is highly prevalent in low- and middle-income countries. It is thought to be a key contributing factor to childhood malnutrition, growth stunting, and diminished oral vaccine responses. Although EE has been shown to be the by-product of a recurrent enteric infection, its full pathophysiology remains unclear. Here, we mapped the cellular and molecular correlates of EE by performing high-throughput, single-cell RNA-sequencing on 33 small intestinal biopsies from 11 adults with EE in Lusaka, Zambia (eight HIV-negative and three HIV-positive), six adults without EE in Boston, United States, and two adults in Durban, South Africa, which we complemented with published data from three additional individuals from the same clinical site. We analyzed previously defined bulk-transcriptomic signatures of reduced villus height and decreased microbial translocation in EE and showed that these signatures may be driven by an increased abundance of surface mucosal cells-a gastric-like subset previously implicated in epithelial repair in the gastrointestinal tract. In addition, we determined cell subsets whose fractional abundances associate with EE severity, small intestinal region, and HIV infection. Furthermore, by comparing duodenal EE samples with those from three control cohorts, we identified dysregulated WNT and MAPK signaling in the EE epithelium and increased proinflammatory cytokine gene expression in a T cell subset highly expressing a transcriptional signature of tissue-resident memory cells in the EE cohort. Together, our work elucidates epithelial and immune correlates of EE and nominates cellular and molecular targets for intervention.
Collapse
Affiliation(s)
- Conner Kummerlowe
- Program in Computational and Systems Biology, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
| | - Simutanyi Mwakamui
- Tropical Gastroenterology and Nutrition group, University of Zambia School of Medicine; Lusaka, Zambia
| | - Travis K. Hughes
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
| | - Nolawit Mulugeta
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
| | - Victor Mudenda
- Tropical Gastroenterology and Nutrition group, University of Zambia School of Medicine; Lusaka, Zambia
| | - Ellen Besa
- Tropical Gastroenterology and Nutrition group, University of Zambia School of Medicine; Lusaka, Zambia
| | - Kanekwa Zyambo
- Tropical Gastroenterology and Nutrition group, University of Zambia School of Medicine; Lusaka, Zambia
| | - Jessica E. S. Shay
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital; Boston, MA, 02114, USA
| | - Ira Fleming
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
| | - Marko Vukovic
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
| | - Ben A. Doran
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital; Boston, MA 02115, USA
| | - Toby P. Aicher
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
| | - Marc H. Wadsworth
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
| | | | - Amiko M. Uchida
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital; Boston, MA 02115, USA
- Cancer Immunology and Virology, Dana Farber Cancer Institute; Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School; Boston MA, 02115, USA
| | - Rabiah Fardoos
- Africa Health Research Institute, Durban, 4001, South Africa
| | | | | | - Ömer H. Yilmaz
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Department of Pathology, MGH, Harvard Medical School, Boston, MA, 02115, USA
| | | | - John J. Garber
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital; Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School; Boston MA, 02115, USA
| | - Jose Ordovas-Montanes
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital; Boston, MA 02115, USA
- Program in Immunology, Harvard Medical School; Boston, MA, 02115, USA
- Harvard Stem Cell Institute; Cambridge, MA, 02138, USA
| | - Zev Gartner
- University of California San Francisco; San Francisco, CA, 94185 USA
| | - Thomas Wallach
- SUNY Downstate Health Sciences University; Department of Pediatrics, Brooklyn, NY, 11203, USA
| | - Alex K. Shalek
- Program in Computational and Systems Biology, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Department of Pathology, MGH, Harvard Medical School, Boston, MA, 02115, USA
- Program in Immunology, Harvard Medical School; Boston, MA, 02115, USA
| | - Paul Kelly
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital; Boston, MA, 02114, USA
- Blizard Institute, Queen Mary University of London; London E1 2AT, United Kingdom
| |
Collapse
|
42
|
Ballas ZK. “Where are they now”? Catching up with the 2017 AAAAI Faculty Development Awardees. J Allergy Clin Immunol 2022; 150:583-584. [DOI: 10.1016/j.jaci.2022.06.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 06/24/2022] [Indexed: 10/16/2022]
|
43
|
Ritzmann F, Lunding LP, Bals R, Wegmann M, Beisswenger C. IL-17 Cytokines and Chronic Lung Diseases. Cells 2022; 11:2132. [PMID: 35883573 PMCID: PMC9318387 DOI: 10.3390/cells11142132] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/27/2022] [Accepted: 07/05/2022] [Indexed: 12/12/2022] Open
Abstract
IL-17 cytokines are expressed by numerous cells (e.g., gamma delta (γδ) T, innate lymphoid (ILC), Th17, epithelial cells). They contribute to the elimination of bacteria through the induction of cytokines and chemokines which mediate the recruitment of inflammatory cells to the site of infection. However, IL-17-driven inflammation also likely promotes the progression of chronic lung diseases, such as chronic obstructive pulmonary disease (COPD), lung cancer, cystic fibrosis, and asthma. In this review, we highlight the role of IL-17 cytokines in chronic lung diseases.
Collapse
Affiliation(s)
- Felix Ritzmann
- Department of Internal Medicine V—Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany; (F.R.); (R.B.)
- Helmholtz Institute for Pharmaceutical Research, 66123 Saarbrücken, Germany
| | - Lars Peter Lunding
- Division of Lung Immunology, Priority Area Asthma and Allergy, Research Center Borstel—Leibniz Lung Center, 23845 Borstel, Germany; (L.P.L.); (M.W.)
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 23845 Borstel, Germany
| | - Robert Bals
- Department of Internal Medicine V—Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany; (F.R.); (R.B.)
- Helmholtz Institute for Pharmaceutical Research, 66123 Saarbrücken, Germany
| | - Michael Wegmann
- Division of Lung Immunology, Priority Area Asthma and Allergy, Research Center Borstel—Leibniz Lung Center, 23845 Borstel, Germany; (L.P.L.); (M.W.)
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 23845 Borstel, Germany
| | - Christoph Beisswenger
- Department of Internal Medicine V—Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany; (F.R.); (R.B.)
| |
Collapse
|
44
|
Ye Q, Bankova LG. Brush cells fine-tune neurogenic inflammation in the airways. J Clin Invest 2022; 132:161439. [PMID: 35775485 PMCID: PMC9246375 DOI: 10.1172/jci161439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Airway epithelial cells, once considered a simple barrier layer, are now recognized as providing an active site for antigen sensing and immune response initiation. Most mucosal sites contain chemosensory epithelial cells, rare and specialized cells gaining recognition for their unique functions in sensing and directing the immune response symphony. In this issue of the JCI, Hollenhorst, Nandigama, et al. demonstrated that tracheal chemosensory brush cells detected bitter-tasting substances, including quorum-sensing molecules (QSMs) generated by pathogenic Pseudomonas aeruginosa. The authors used various techniques, including genetic deletion of brush cells, genetic manipulation of brush cell signaling, deletion of sensory neurons, in vivo imaging, and infection models with P. aeruginosa, to show that QSMs increased vascular permeability and innate immune cell influx into the trachea. These findings link the recognition of bacterial QSMs to the innate immune response in the airways, with translational implications for airway inflammation and infectious pathology.
Collapse
|
45
|
Inclan-Rico JM, Rossi HL, Herbert DR. "Every cell is an immune cell; contributions of non-hematopoietic cells to anti-helminth immunity". Mucosal Immunol 2022; 15:1199-1211. [PMID: 35538230 PMCID: PMC9646929 DOI: 10.1038/s41385-022-00518-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/04/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023]
Abstract
Helminths are remarkably successful parasites that can invade various mammalian hosts and establish chronic infections that can go unnoticed for years despite causing severe tissue damage. To complete their life cycles, helminths migrate through multiple barrier sites that are densely populated by a complex array of hematopoietic and non-hematopoietic cells. While it is clear that type 2 cytokine responses elicited by immune cells promote worm clearance and tissue healing, the actions of non-hematopoietic cells are increasingly recognized as initiators, effectors and regulators of anti-helminth immunity. This review will highlight the collective actions of specialized epithelial cells, stromal niches, stem, muscle and neuroendocrine cells as well as peripheral neurons in the detection and elimination of helminths at mucosal sites. Studies dissecting the interactions between immune and non-hematopoietic cells will truly provide a better understanding of the mechanisms that ensure homeostasis in the context of helminth infections.
Collapse
Affiliation(s)
- Juan M Inclan-Rico
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Heather L Rossi
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - De'Broski R Herbert
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
46
|
New insights into tuft cell formation: Implications for structure–function relationships. Curr Opin Cell Biol 2022; 76:102082. [DOI: 10.1016/j.ceb.2022.102082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/18/2022] [Accepted: 03/24/2022] [Indexed: 11/18/2022]
|
47
|
Jakwerth CA, Ordovas-Montanes J, Blank S, Schmidt-Weber CB, Zissler UM. Role of Respiratory Epithelial Cells in Allergic Diseases. Cells 2022; 11:1387. [PMID: 35563693 PMCID: PMC9105716 DOI: 10.3390/cells11091387] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/11/2022] [Accepted: 04/19/2022] [Indexed: 02/07/2023] Open
Abstract
The airway epithelium provides the first line of defense to the surrounding environment. However, dysfunctions of this physical barrier are frequently observed in allergic diseases, which are tightly connected with pro- or anti-inflammatory processes. When the epithelial cells are confronted with allergens or pathogens, specific response mechanisms are set in motion, which in homeostasis, lead to the elimination of the invaders and leave permanent traces on the respiratory epithelium. However, allergens can also cause damage in the sensitized organism, which can be ascribed to the excessive immune reactions. The tight interaction of epithelial cells of the upper and lower airways with local and systemic immune cells can leave an imprint that may mirror the pathophysiology. The interaction with effector T cells, along with the macrophages, play an important role in this response, as reflected in the gene expression profiles (transcriptomes) of the epithelial cells, as well as in the secretory pattern (secretomes). Further, the storage of information from past exposures as memories within discrete cell types may allow a tissue to inform and fundamentally alter its future responses. Recently, several lines of evidence have highlighted the contributions from myeloid cells, lymphoid cells, stromal cells, mast cells, and epithelial cells to the emerging concepts of inflammatory memory and trained immunity.
Collapse
Affiliation(s)
- Constanze A. Jakwerth
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health, Germany, Member of the German Center for Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, 80802 Munich, Germany; (C.A.J.); (S.B.); (C.B.S.-W.)
| | - Jose Ordovas-Montanes
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA 02115, USA;
- Program in Immunology, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Simon Blank
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health, Germany, Member of the German Center for Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, 80802 Munich, Germany; (C.A.J.); (S.B.); (C.B.S.-W.)
| | - Carsten B. Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health, Germany, Member of the German Center for Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, 80802 Munich, Germany; (C.A.J.); (S.B.); (C.B.S.-W.)
| | - Ulrich M. Zissler
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health, Germany, Member of the German Center for Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, 80802 Munich, Germany; (C.A.J.); (S.B.); (C.B.S.-W.)
| |
Collapse
|
48
|
Hawerkamp HC, Fallon PG. Expelliarmus helminthus! Harry Helminth and the Goblet of Alarmins. Immunity 2022; 55:575-577. [PMID: 35417668 DOI: 10.1016/j.immuni.2022.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Epithelial cells (tuft and goblet cells) interact with immune cells on the "inside" while secreting effector molecules into the topological "outside." In this issue of Immunity, Zhao et al. investigate an interleukin-33 (IL-33) secretion mechanism in goblet cells dependent on O-GlcNAcylation and gasdermin pores facilitating worm expulsion.
Collapse
Affiliation(s)
- Heike C Hawerkamp
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| | - Padraic G Fallon
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
49
|
Vacca F, Le Gros G. Tissue-specific immunity in helminth infections. Mucosal Immunol 2022; 15:1212-1223. [PMID: 35680972 PMCID: PMC9178325 DOI: 10.1038/s41385-022-00531-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/25/2022] [Accepted: 05/16/2022] [Indexed: 02/04/2023]
Abstract
A characteristic feature of host responses to helminth infections is the development of profound systemic and tissue-localised Type 2 immune responses that play critical roles in immunity, tissue repair and tolerance of the parasite at tissue sites. These same Type 2 responses are also seen in the tissue-associated immune-pathologies seen in asthma, atopic dermatitis and many forms of allergies. The recent identification of new subtypes of immune cells and cytokine pathways that influence both immune and non-immune cells and tissues creates the opportunity for reviewing helminth parasite-host responses in the context of tissue specific immunity. This review focuses on the new discoveries of the cells and cytokines involved in tissue specific immune responses to helminths and how these contribute to host immunity against helminth infection and allow the host to accommodate the presence of parasites when they cannot be eliminated.
Collapse
Affiliation(s)
- Francesco Vacca
- grid.250086.90000 0001 0740 0291Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Graham Le Gros
- grid.250086.90000 0001 0740 0291Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
50
|
Trained immunity in type 2 immune responses. Mucosal Immunol 2022; 15:1158-1169. [PMID: 36065058 PMCID: PMC9705254 DOI: 10.1038/s41385-022-00557-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/27/2022] [Accepted: 08/08/2022] [Indexed: 02/04/2023]
Abstract
Immunological memory of innate immune cells, also termed "trained immunity", allows for cross-protection against distinct pathogens, but may also drive chronic inflammation. Recent studies have shown that memory responses associated with type 2 immunity do not solely rely on adaptive immune cells, such as T- and B cells, but also involve the innate immune system and epithelial cells. Memory responses have been described for monocytes, macrophages and airway epithelial cells of asthmatic patients as well as for macrophages and group 2 innate lymphoid cells (ILC2) from allergen-sensitized or helminth-infected mice. The metabolic and epigenetic mechanisms that mediate allergen- or helminth-induced reprogramming of innate immune cells are only beginning to be uncovered. Trained immunity has been implicated in helminth-driven immune regulation and allergen-specific immunotherapy, suggesting its exploitation in future therapies. Here, we discuss recent advances and key remaining questions regarding the mechanisms and functions of trained type 2 immunity in infection and inflammation.
Collapse
|