1
|
Fu L, Yokus B, Gao B, Pacher P. An Update on IL-22 Therapies in Alcohol-Associated Liver Disease and Beyond. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00117-8. [PMID: 40254130 DOI: 10.1016/j.ajpath.2025.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/22/2025]
Abstract
Excessive alcohol consumption drives the development of alcohol-associated liver disease (ALD), including steatohepatitis, cirrhosis, and hepatocellular carcinoma, and its associated complications, such as hepatorenal syndrome. Hepatocyte death, inflammation, and impaired liver regeneration are key processes implicated in the pathogenesis and progression of ALD. Despite extensive research, therapeutic options for ALD remain limited. IL-22 has emerged as a promising therapeutic target because of its hepatoprotective properties mediated through the activation of the STAT3 signaling pathway. IL-22 enhances hepatocyte survival by mitigating apoptosis, oxidative stress, and inflammation while simultaneously promoting liver regeneration through the proliferation of hepatocytes and hepatic progenitor cells and the up-regulation of growth factors. Additionally, IL-22 exerts protective effects on epithelial cells in various organs affected by ALD and its associated complications. Studies from preclinical models and early-phase clinical trials of IL-22 agonists, such as F-652 and UTTR1147A, have shown favorable safety profiles, good tolerability, and encouraging efficacy in reducing liver injury and promoting regeneration. However, the heterogeneity and multifactorial nature of ALD present ongoing challenges. Further research is needed to optimize IL-22-based therapies and clarify their roles within a comprehensive approach to ALD management. This review summarizes the current understanding of IL-22 biology and its role in ALD pathophysiology and ALD-associated complications along with therapeutic application of IL-22, potential benefits, and limitations.
Collapse
Affiliation(s)
- Lihong Fu
- Laboratory of Cardiovascular Physiology and Tissue Injury, NIH/National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland
| | - Burhan Yokus
- Laboratory of Cardiovascular Physiology and Tissue Injury, NIH/National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland
| | - Bin Gao
- Laboratory of Liver Diseases, NIH/National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland.
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, NIH/National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland.
| |
Collapse
|
2
|
Soliman GA, Alamri MA, Abdel-Rahman RF, Elbaset MA, Ogaly HA, Abdel-Kader MS. Tephrosia purpurea, with (-)-Pseudosemiglabrin as the Major Constituent, Alleviates Severe Acute Pancreatitis-Mediated Acute Lung Injury by Modulating HMGB1 and IL-22. Int J Mol Sci 2025; 26:2572. [PMID: 40141214 PMCID: PMC11942157 DOI: 10.3390/ijms26062572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 03/02/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Ischemia-reperfusion (IR) injury is a major cause of multiple organ failure. The purpose of this study was to look into the role of Tephrosia purpurea (TEP) and its active constituent pseudosemiglabrin (PS) in alleviating severe acute pancreatitis and its associated acute lung injury. We established a rat pancreatic IR model, and the rats were treated with TEP (200 mg/kg and 400 mg/kg) and PS (20 and 40 mg/kg), in addition to the IR control and sham groups. The results showed that the respiratory parameters, including inspiratory time (Ti), expiratory time (Te), duration (Dr), and respiratory rate (RR), were comparable among all groups, while peak inspiratory flow (PIF), forced vital capacity (FVC), and forced expiratory volume at 0.1 s (FEV0.1) were significantly impaired. Notably, PS at 40 mg/kg showed normal PIF, FVC, and FEV0.1/FVC compared to the IR group, indicating an improved lung function. Additionally, TEP and PS showed protective effects on pancreatic and lung tissues compared to the IR control group, with the following effects: alleviating pathological damage; reducing serum levels of trypsinogen activation peptide (TAP), lipase, and amylase; decreasing oxidative stress markers such as MDA and MPO; restoring antioxidant enzyme activity (GPx); suppressing inflammatory markers TNF-α, IL-6, and NF-κB; downregulating HMGB1 gene in pancreatic tissue; and upregulating the IL-22 gene in lung tissues. In conclusion, the obtained findings demonstrate that oral supplementation of TEP and PS to rats with pancreatic IR alleviates pancreatic and lung injuries by reducing oxidative stress and modulating inflammatory processes, which offers an attractive therapeutic option for severe acute pancreatitis and its associated acute lung injury.
Collapse
Affiliation(s)
- Gamal A. Soliman
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (G.A.S.); (M.A.A.)
| | - Mohammed A. Alamri
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (G.A.S.); (M.A.A.)
| | - Rehab F. Abdel-Rahman
- Department of Pharmacology, National Research Centre, Giza 12622, Egypt; (R.F.A.-R.); (M.A.E.)
| | - Marawan A. Elbaset
- Department of Pharmacology, National Research Centre, Giza 12622, Egypt; (R.F.A.-R.); (M.A.E.)
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hanan A. Ogaly
- Department of Biochemistry, College of Veterinary Medicine, Cairo University, Giza 12613, Egypt;
| | - Maged S. Abdel-Kader
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| |
Collapse
|
3
|
Stadler SV, von Garnier C, Ubags ND. Post-viral lung diseases: the microbiota as a key player. ERJ Open Res 2025; 11:00560-2024. [PMID: 40196711 PMCID: PMC11973713 DOI: 10.1183/23120541.00560-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/08/2024] [Indexed: 04/09/2025] Open
Abstract
Viral infections of the respiratory tract can lead to chronic lung injury through immunopathological mechanisms that remain unclear. Communities of commensal bacteria colonising the respiratory tract, known as the respiratory tract microbiota, are altered in viral infections, which can contribute to inflammation, lung epithelial damage and subsequent development of lung disease. Emerging evidence on post-viral lung injury suggests an interplay between viral infections, immune responses and airway microbiota composition in the development of viral-induced lung diseases. In this review, we present the clinical characteristics of post-viral lung injury, along with the underlying immunopathological mechanisms and host-bacteria interactions, with a focus on influenza virus, respiratory syncytial virus and coronaviruses. Additionally, considering the important role of the airway microbiota in viral-induced pulmonary sequelae, we suggest key areas for future research on respiratory microbiota involvement in the development of post-viral lung diseases.
Collapse
Affiliation(s)
- Sabine V. Stadler
- Division of Pulmonary Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Christophe von Garnier
- Division of Pulmonary Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Niki D. Ubags
- Division of Pulmonary Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
4
|
Threatt AN, White J, Klepper N, Brier Z, Dean LS, Ibarra A, Harris M, Jones K, Wahl MJL, Barahona M, Oyewole EO, Pauly M, Moreno JA, Nordgren TM. Aspirin-triggered resolvin D1 modulates pulmonary and neurological inflammation in an IL-22 knock-out organic dust exposure mouse model. Front Immunol 2024; 15:1495581. [PMID: 39776904 PMCID: PMC11705093 DOI: 10.3389/fimmu.2024.1495581] [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: 09/12/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Agriculture dust contains many organic immunogenic compounds, and organic dust exposure is strongly associated with the development of immune-mediated chronic pulmonary diseases such as chronic obstructive pulmonary disease (COPD). Chronic organic dust exposure from agriculture sources induces chronic lung inflammatory diseases and organic dust exposure has recently been linked to an increased risk of developing dementia. The cytokine interleukin-22 (IL-22) has been established as an important mediator in the resolution and repair of lung tissues. The omega-3 fatty acid metabolite aspirin-triggered Resolvin D1 (AT-RvD1) has shown efficacy in modulating the immune response in both pulmonary and neurological inflammation but has not been explored as a therapeutic in organic dust exposure-induced neuroinflammation. Investigating the link between IL-22 and AT-RvD1 may help in developing effective therapies for these immune-mediated diseases. We aimed to investigate the link between organic dust exposure and neuroinflammation, the role of IL-22 in the pulmonary and neurological immune response to organic dust exposure, and the immune-modulating therapeutic applications of AT-RvD1 in an IL-22 knock-out mouse model of organic dust exposure. C57BL/6J (WT) and IL-22 knock-out (KO) mice were repetitively exposed to aqueous agriculture organic dust extract (DE) 5 days per week for 3 weeks (15 total instillations) and treated with AT-RvD1 either once per week (3 total injections) or 5 times per week (15 total injections) for 3 weeks and allowed to recover for 3 days. We observed a significant pulmonary and neurological immune response to DE characterized by the development of inducible bronchus associated lymphoid tissue in the lung and gliosis in the frontal areas of the brain. We also observed that IL-22 knock-out increased pulmonary and neurological inflammation severity. Animals exposed to DE and treated with AT-RvD1 displayed reduced lung pathology severity and gliosis. Our data demonstrate that DE exposure contributes to neurological inflammation and that IL-22 is crucial to effective tissue repair processes. Our data further suggest that AT-RvD1 may have potential as a novel therapeutic for organic dust exposure-induced, immune-mediated pulmonary and neurological inflammation, improving outcomes of those with these diseases.
Collapse
Affiliation(s)
- Alissa N. Threatt
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Jade White
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Biology, College of Natural Sciences, Colorado State University, Fort Collins, CO, United States
| | - Nathan Klepper
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Animal Sciences, College of Agricultural Sciences, Colorado State University, Fort Collins, CO, United States
| | - Zachary Brier
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Logan S. Dean
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, United States
| | - Ash Ibarra
- Department of Chemistry, College of Natural Sciences, Colorado State University, Fort Collins, CO, United States
| | - Macallister Harris
- Experimental Pathology Facility, Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Kaylee Jones
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Maëlis J. L. Wahl
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, CO, United States
| | - Melea Barahona
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, United States
| | - Emmanuel O. Oyewole
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Morgan Pauly
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, CO, United States
| | - Julie A. Moreno
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Brain Research Center, Colorado State University, Fort Collins, CO, United States
| | - Tara M. Nordgren
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
5
|
Dean LS, Threatt AN, Jones K, Oyewole EO, Pauly M, Wahl M, Barahona M, Reiter RW, Nordgren TM. I don't know about you, but I'm feeling IL-22. Cytokine Growth Factor Rev 2024; 80:1-11. [PMID: 39537498 DOI: 10.1016/j.cytogfr.2024.11.001] [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: 10/01/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Defense of the human body against damaging and pathogenic insults is a heavily regulated affair. A primary mechanism of defense at sites of insult are soluble mediators whose defensive maneuvers increase barrier integrity and promote pro-reparative and resolution processes. IL-22 is a cytokine in the IL-10 cytokine family that has garnered increased attention in recent years due to its intimate link in promoting resolution of inflammatory insults, while simultaneously being over expressed in certain fibrotic and chronic inflammatory-skewed diseases. The spatial action of IL-22 centers around the barrier sites of the body, including the skin, lungs, and gut mucosa. As such, a detailed understanding of the role of this cytokine, the producers and responders, and the diseases resulting from over- or under-expression have prominent impacts on a variety of disease outcomes. Herein we present a comprehensive review of IL-22; from historical perspectives and initial discovery, as well as more recent data that dramatically expands on the cellular sources and impact of this cytokine. We aim to showcase the duality of IL-22 and highlight addressable gaps in the field of IL-22 crosstalk and impacts at the ever-important mucosal and tissue barrier sites.
Collapse
Affiliation(s)
- Logan S Dean
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO 80521, United States; Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80521, United States
| | - Alissa N Threatt
- Toxicology Graduate Program, Colorado State University, Fort Collins, CO 80521, United States; Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80521, United States
| | - Kaylee Jones
- Toxicology Graduate Program, Colorado State University, Fort Collins, CO 80521, United States; Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80521, United States
| | - Emmanuel O Oyewole
- Toxicology Graduate Program, Colorado State University, Fort Collins, CO 80521, United States; Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80521, United States
| | - Morgan Pauly
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80521, United States
| | - Maëlis Wahl
- Department of Biochemistry and Molecular Biology, Colorado State University, CO 80521, United States
| | - Melea Barahona
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO 80521, United States; Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80521, United States
| | - Rose W Reiter
- Department of Molecular, Cellular, and Integrative Neuroscience, Colorado State University, CO 80521, United States
| | - Tara M Nordgren
- Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO 80521, United States; Toxicology Graduate Program, Colorado State University, Fort Collins, CO 80521, United States; Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80521, United States.
| |
Collapse
|
6
|
Savin IA, Sen’kova AV, Goncharova EP, Zenkova MA, Markov AV. Novel Core Gene Signature Associated with Inflammation-to-Metaplasia Transition in Influenza A Virus-Infected Lungs. Int J Mol Sci 2024; 25:11958. [PMID: 39596028 PMCID: PMC11594146 DOI: 10.3390/ijms252211958] [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: 10/09/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Respiratory infections caused by RNA viruses are a major contributor to respiratory disease due to their ability to cause annual epidemics with profound public health implications. Influenza A virus (IAV) infection can affect a variety of host signaling pathways that initiate tissue regeneration with hyperplastic and/or dysplastic changes in the lungs. Although these changes are involved in lung recovery after IAV infection, in some cases, they can lead to serious respiratory failure. Despite being ubiquitously observed, there are limited data on the regulation of long-term recovery from IAV infection leading to normal or dysplastic repair represented by inflammation-to-metaplasia transition in mice or humans. To address this knowledge gap, we used integrative bioinformatics analysis with further verification in vivo to elucidate the dynamic molecular changes in IAV-infected murine lung tissue and identified the core genes (Birc5, Cdca3, Plk1, Tpx2, Prc1. Rrm2, Nusap1, Spag5, Top2a, Mcm5) and transcription factors (E2F1, E2F4, NF-YA, NF-YB, NF-YC) involved in persistent lung injury and regeneration processes, which may serve as gene signatures reflecting the long-term effects of IAV proliferation on the lung. Further analysis of the identified core genes revealed their involvement not only in IAV infection but also in COVID-19 and lung neoplasm development, suggesting their potential role as biomarkers of severe lung disease and its complications represented by abnormal epithelial proliferation and oncotransformation.
Collapse
|
7
|
Bochnia-Bueno L, Coelho GM, Cataneo AHD, Zanluca C, Ferreira LH, Cavalcanti LPDG, Clementino MADF, Yaochite JNU, Dos Santos HG, Nogueira MB, Duarte Dos Santos CN, Raboni SM. Assessment of immune responses to a Comirnaty® booster following CoronaVac® vaccination in healthcare workers. Mem Inst Oswaldo Cruz 2024; 119:e230239. [PMID: 39258622 PMCID: PMC11385826 DOI: 10.1590/0074-02760230239] [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: 12/20/2023] [Accepted: 06/13/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND The immunological response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and immunisation is variable. OBJECTIVES To describe the humoral immune response by correlating IgA and IgG antibodies with NAbs titration following CoronaVac® immunisation and an mRNA (Comirnaty®) booster among healthcare workers (HCWs) and to compare the cytokine and interleukin profiles between HCWs vaccinated with CoronaVac and coronavirus disease 2019 (COVID-19) infected patients. METHODS Samples from 133 HCWs collected at 20 (T1) and 90 (T2) days after CoronaVac immunisation and 15 (T3) days after a booster dose with the Comirnaty vaccine were analysed for IgA and IgG EIA and neutralisation assay. Cytokine levels from vaccinated individuals at T1 day and COVID-19 patients were compared. FINDINGS Neutralising antibodies (NAbs) were observed in 81.7% of participants at T1, but only 49.2% maintained detectable NAbs after 90 days. The booster dose increased NAbs response in all participants. The cytokines with the highest levels post-vaccination were IL-6 and MCP-1. The MCP-1, IL-18, and IFN- γ levels were higher in COVID-19 patients than in vaccinated HCWs, while IL-22 levels increased in the vaccinated HCWs group. MAIN CONCLUSIONS The neutralisation titres in the T2 samples decreased, and antibody levels detected at T2 showed a more significant reduction than the neutralisation. The higher IL-22 expression in immunised individuals compared to those with COVID-19 suggests that IL-22 may be beneficial in protecting against severe disease.
Collapse
Affiliation(s)
- Lucas Bochnia-Bueno
- Universidade Federal do Paraná, Laboratório de Virologia, Curitiba, PR, Brasil
- Universidade Federal do Paraná, Programa de Pós-Graduação em Microbiologia, Parasitologia e Patologia, Curitiba, PR, Brasil
| | - Gabriela Mattoso Coelho
- Fundação Oswaldo Cruz-Fiocruz, Instituto Carlos Chagas, Laboratório de Virologia Molecular, Curitiba, PR, Brasil
| | | | - Camila Zanluca
- Fundação Oswaldo Cruz-Fiocruz, Instituto Carlos Chagas, Laboratório de Virologia Molecular, Curitiba, PR, Brasil
| | - Laura Holtman Ferreira
- Universidade Federal do Paraná, Laboratório de Virologia, Curitiba, PR, Brasil
- Universidade Federal do Paraná, Programa de Pós-Graduação em Microbiologia, Parasitologia e Patologia, Curitiba, PR, Brasil
| | | | | | - Juliana Navarro Ueda Yaochite
- Universidade Federal do Ceará, Faculdade de Farmácia, Odontologia e Enfermagem, Departamento de Análises Clínicas e Toxicologia, Fortaleza, CE, Brasil
| | | | - Meri Bordignon Nogueira
- Universidade Federal do Paraná, Laboratório de Virologia, Curitiba, PR, Brasil
- Universidade Federal do Paraná, Programa de Pós-Graduação em Microbiologia, Parasitologia e Patologia, Curitiba, PR, Brasil
| | | | - Sonia Mara Raboni
- Universidade Federal do Paraná, Laboratório de Virologia, Curitiba, PR, Brasil
- Universidade Federal do Paraná, Programa de Pós-Graduação em Microbiologia, Parasitologia e Patologia, Curitiba, PR, Brasil
| |
Collapse
|
8
|
Guo C, Boulant S, Stanifer ML. The Role of Interleukin-22 in Controlling Virus Infections at Mucosal Surfaces. J Interferon Cytokine Res 2024; 44:349-354. [PMID: 38868897 DOI: 10.1089/jir.2024.0097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Affiliation(s)
- Cuncai Guo
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Heidelberg, Germany
| | - Steeve Boulant
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Heidelberg, Germany
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Megan Lynn Stanifer
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
9
|
Huckestein BR, Antos D, Manni ML, Zeng K, Miller LM, Parenteau KL, Gelhaus SL, Mullett SJ, Shoemaker JE, Alcorn JF. Sex-based differences in persistent lung inflammation following influenza infection of juvenile outbred mice. Am J Physiol Lung Cell Mol Physiol 2024; 327:L189-L202. [PMID: 38810239 PMCID: PMC11687968 DOI: 10.1152/ajplung.00407.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/16/2024] [Accepted: 05/24/2024] [Indexed: 05/31/2024] Open
Abstract
Children are susceptible to influenza infections and can experience severe disease presentation due to a lack of or limited pre-existing immunity. Despite the disproportionate impact influenza has on this population, there is a lack of focus on pediatric influenza research, particularly when it comes to identifying the pathogenesis of long-term outcomes that persist beyond the point of viral clearance. In this study, juvenile outbred male and female mice were infected with influenza and analyzed following viral clearance to determine how sex impacts the persistent inflammatory responses to influenza. It was found that females maintained a broader cytokine response in the lung following clearance of influenza, with innate, type I and type II cytokine signatures in almost all mice. Males, on the other hand, had higher levels of IL-6 and other macrophage-related cytokines, but no evidence of a type I or type II response. The immune landscape was similar in the lungs between males and females postinfection, but males had a higher regulatory T cell to TH1 ratio compared with female mice. Cytokine production positively correlated with the frequency of TH1 cells and exudate macrophages, as well as the number of cells in the bronchoalveolar lavage fluid. Furthermore, female lungs were enriched for metabolites involved in the glycolytic pathway, suggesting glycolysis is higher in female lungs compared with males after viral clearance. These data suggest juvenile female mice have persistent and excessive lung inflammation beyond the point of viral clearance, whereas juvenile males had a more immunosuppressive phenotype.NEW & NOTEWORTHY This study identifies sex-based differences in persistent lung inflammation following influenza infection in an outbred, juvenile animal model of pediatric infection. These findings indicate the importance of considering sex and age as variable in infectious disease research.
Collapse
Affiliation(s)
- Brydie R Huckestein
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Danielle Antos
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Michelle L Manni
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Kelly Zeng
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Leigh M Miller
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Kristen L Parenteau
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Stacy L Gelhaus
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Steven J Mullett
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Jason E Shoemaker
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - John F Alcorn
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
10
|
Pillar A, Ali MK. IL-22 Binding Protein/IL-22 Axis in Regulating Acute Lung Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:335-337. [PMID: 38199431 DOI: 10.1016/j.ajpath.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024]
Affiliation(s)
- Amber Pillar
- School of Biomedical Sciences and Pharmacy, University of Newcastle and The Immune Health Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Md Khadem Ali
- Pre-Professional Health Academic Program, California State University East Bay, Hayward, California.
| |
Collapse
|
11
|
Zhang Z, Chakawa MB, Galeas-Pena M, Frydman JA, Allen MJ, Jones M, Pociask D. IL-22 Binding Protein Controls IL-22-Driven Bleomycin-Induced Lung Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:338-352. [PMID: 38101567 PMCID: PMC10913761 DOI: 10.1016/j.ajpath.2023.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 10/02/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023]
Abstract
The high mortality rates of acute lung injury and acute respiratory distress syndrome challenge the field to identify biomarkers and factors that can be exploited for therapeutic approaches. IL-22 is a cytokine that has antibacterial and reparative properties in the lung. However, it also can exacerbate inflammation and requires tight control by the extracellular inhibitory protein known as IL-22 binding protein (IL-22BP) (Il22ra2). This study showed the necessity of IL-22BP in controlling and preventing acute lung injury using IL-22BP knockout mice (Il22ra2-/-) in the bleomycin model of acute lung injury/acute respiratory distress syndrome. Il22ra2-/- mice had greater sensitivity (weight loss and death) and pulmonary inflammation in the acute phase (first 7 days) of the injury compared with wild-type C57Bl/6 controls. The inflammation was driven by excess IL-22 production, inducing the influx of pathogenic IL-17A+ γδ T cells to the lung. Interestingly, this inflammation was initiated in part by the noncanonical IL-22 signaling to macrophages, which express the IL-22 receptor (Il22ra1) in vivo after bleomycin challenge. This study further showed that IL-22 receptor alpha-1+ macrophages can be stimulated by IL-22 to produce a number of IL-17-inducing cytokines such as IL-1β, IL-6, and transforming growth factor-β1. Together, the results suggest that IL-22BP prevents IL-22 signaling to macrophages and reduces bleomycin-mediated lung injury.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Medicine, Pulmonary Diseases, Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Mazvita B Chakawa
- Department of Medicine, Pulmonary Diseases, Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Michelle Galeas-Pena
- Department of Medicine, Pulmonary Diseases, Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Joshua A Frydman
- Department of Medicine, Pulmonary Diseases, Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Michaela J Allen
- Department of Medicine, Pulmonary Diseases, Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - MaryJane Jones
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Derek Pociask
- Department of Medicine, Pulmonary Diseases, Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, Louisiana.
| |
Collapse
|
12
|
Guo L, Bao W, Yang S, Liu Y, Lyu J, Wang T, Lu Y, Li H, Zhu H, Chen D. Rhei Radix et Rhizoma in Xuanbai-Chengqi decoction strengthens the intestinal barrier function and promotes lung barrier repair in preventing severe viral pneumonia induced by influenza A virus. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117231. [PMID: 37783404 DOI: 10.1016/j.jep.2023.117231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/22/2023] [Accepted: 09/24/2023] [Indexed: 10/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xuanbai-Chengqi decoction (XCD) is a traditional prescription for treating multiple organ injuries, which has been used to manage pneumonia caused by various pathogens. However, the effects of XCD on repairing pulmonary/intestinal barrier damage remain unclear, and there is a need to understand the compatibility mechanism of rhubarb. AIM OF THE STUDY This work aims to investigate the protective effect and mechanism of XCD on the pulmonary/intestinal barrier guided by the theory of "gut-lung concurrent treatment". Moreover, we elucidate the compatibility mechanism of rhubarb in XCD. MATERIALS AND METHODS An H1N1 virus-infected mouse model was adopted to investigate the reparative effects of XCD on the lung-intestinal barrier by assessing lung-intestinal permeability. Additionally, the characterization of type I alveolar epithelial cells (AT1) and type II alveolar epithelial cells (AT2) was performed to evaluate the damage to the alveolar epithelial barrier. The specific barrier-protective mechanisms of XCD were elucidated by detecting tight junction proteins and the epithelial cell repair factor IL-22. The role of rhubarb in XCD to pneumonia treatment was investigated through lung tissue transcriptome sequencing and flow cytometry. RESULTS XCD significantly improved lung tissue edema, inflammation, and alveolar epithelial barrier damage by regulating IL-6, IL-10, and IL-22, which, could further improve pulmonary barrier permeability when combined with the protection of alveolar epithelial cells (AT1 and AT2) as well as inhibition of H1N1 virus replication. Simultaneously, XCD significantly reduced intestinal inflammation and barrier damage by regulating IL-6, IL-1β, and tight junction protein levels (Claudin-1 and ZO-1), improving intestinal barrier permeability. The role of rhubarb in the treatment of pneumonia is clarified for the first time. In the progression of severe pneumonia, rhubarb can significantly protect the intestinal barrier, promote the repair of AT2 cells, and inhibit the accumulation of CD11b+Ly6Gvariable aberrant neutrophils by regulating the S100A8 protein. CONCLUSION In summary, our findings suggest that rhubarb in XCD plays a critical role in protecting intestinal barrier function and promoting lung barrier repair in preventing severe viral pneumonia caused by influenza A virus.
Collapse
Affiliation(s)
- Linfeng Guo
- Department of Natural Medicine, School of Pharmacy, Fudan University, 3728# Jinke Rd., Pudong District, Shanghai, 201203, PR China
| | - Weilian Bao
- Department of Natural Medicine, School of Pharmacy, Fudan University, 3728# Jinke Rd., Pudong District, Shanghai, 201203, PR China
| | - Shuiyuan Yang
- Department of Natural Medicine, School of Pharmacy, Fudan University, 3728# Jinke Rd., Pudong District, Shanghai, 201203, PR China
| | - Yang Liu
- Department of Natural Medicine, School of Pharmacy, Fudan University, 3728# Jinke Rd., Pudong District, Shanghai, 201203, PR China
| | - Jiaren Lyu
- Department of Natural Medicine, School of Pharmacy, Fudan University, 3728# Jinke Rd., Pudong District, Shanghai, 201203, PR China
| | - Ting Wang
- Department of Biological Medicines, Shanghai Engineering Research Center of ImmunoTherapeutics, School of Pharmacy, Fudan University, 3728# Jinke Rd., Pudong, District, Shanghai, 201203, PR China
| | - Yan Lu
- Department of Natural Medicine, School of Pharmacy, Fudan University, 3728# Jinke Rd., Pudong District, Shanghai, 201203, PR China
| | - Hong Li
- Department of Pharmacology, School of Pharmacy, Fudan University, 3728# Jinke Rd., Pudong, Shanghai, 201203, PR China
| | - Haiyan Zhu
- Department of Biological Medicines, Shanghai Engineering Research Center of ImmunoTherapeutics, School of Pharmacy, Fudan University, 3728# Jinke Rd., Pudong, District, Shanghai, 201203, PR China.
| | - Daofeng Chen
- Department of Natural Medicine, School of Pharmacy, Fudan University, 3728# Jinke Rd., Pudong District, Shanghai, 201203, PR China.
| |
Collapse
|
13
|
Hsu CY, Faisal Mutee A, Porras S, Pineda I, Ahmed Mustafa M, J Saadh M, Adil M, H A Z. Amphiregulin in infectious diseases: Role, mechanism, and potential therapeutic targets. Microb Pathog 2024; 186:106463. [PMID: 38036111 DOI: 10.1016/j.micpath.2023.106463] [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: 10/07/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
Amphiregulin (AREG) serves as a ligand for the epidermal growth factor receptor (EGFR) and is involved in vital biological functions, including inflammatory responses, tissue regeneration, and immune system function. Upon interaction with the EGFR, AREG initiates a series of signaling cascades necessary for several physiological activities, such as metabolism, cell cycle regulation, and cellular proliferation. Recent findings have provided evidence for the substantial role of AREG in maintaining the equilibrium of homeostasis in damaged tissues and preserving epithelial cell structure in the context of viral infections affecting the lungs. The development of resistance to influenza virus infection depends on the presence of type 1 cytokine responses. Following the eradication of the pathogen, the lungs are subsequently colonized by several cell types that are linked with type 2 immune responses. These cells contribute to the process of repairing and resolving the tissue injury and inflammation caused by infections. Following influenza infection, the activation of AREG promotes the regeneration of bronchial epithelial cells, enhancing the tissue's structural integrity and increasing the survival rate of infected mice. In the same manner, mice afflicted with influenza experience rapid mortality due to a subsequent bacterial infection in the pulmonary region when both bacterial and viral infections manifest concurrently inside the same host. The involvement of AREG in bacterial infections has been demonstrated. The gene AREG experiences increased transcriptional activity inside host cells in response to bacterial infections caused by pathogens such as Escherichia coli and Neisseria gonorrhea. In addition, AREG has been extensively studied as a mitogenic stimulus in epithelial cell layers. Consequently, it is regarded as a prospective contender that might potentially contribute to the observed epithelial cell reactions in helminth infection. Consistent with this finding, mice that lack the AREG gene exhibit a delay in the eradication of the intestinal parasite Trichuris muris. The observed delay is associated with a reduction in the proliferation rate of colonic epithelial cells compared to the infected animals in the control group. The aforementioned findings indicate that AREG plays a pivotal role in facilitating the activation of defensive mechanisms inside the epithelial cells of the intestinal tissue. The precise cellular sources of AREG in this specific context have not yet been determined. However, it is evident that the increased proliferation of the epithelial cell layer in infected mice is reliant on CD4+ T cells. The significance of this finding lies in its demonstration of the crucial role played by the interaction between immunological and epithelial cells in regulating the AREG-EGFR pathway. Additional research is necessary to delve into the cellular origins and signaling mechanisms that govern the synthesis of AREG and its tissue-protective properties, independent of infection.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan City 71710, Taiwan
| | | | - Sandra Porras
- Facultad de Mecánica, Escuela Superior Politécnica de Chimborazo (ESPOCH), Panamericana Sur km 1 1/2, Riobamba, 060155, Ecuador
| | - Indira Pineda
- Facultad de Salud Pública, Escuela Superior Politécnica de Chimborazo (ESPOCH), Panamericana Sur km 1 1/2, Riobamba, 060155, Ecuador
| | - Mohammed Ahmed Mustafa
- Department of Medical Laboratory Technology, Imam Jaafar AL-Sadiq University, Iraq; Department of Pathological Analyzes, College of Applied Sciences, University of Samarra, Iraq.
| | - Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan; Applied Science Research Center, Applied Science Private University, Amman, Jordan
| | | | - Zainab H A
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| |
Collapse
|
14
|
Gopal R, Tutuncuoglu E, Bakalov V, Wasserloos K, Li H, Lemley D, DeVito LJ, Constantinesco NJ, Reed DS, McHugh KJ, Chinnappan B, Andreas AR, Maloy A, Bain D, Alcorn JF, Pitt BR, Kaynar AM. Zinc deficiency enhances sensitivity to influenza A associated bacterial pneumonia in mice. Physiol Rep 2024; 12:e15902. [PMID: 38163670 PMCID: PMC10758336 DOI: 10.14814/phy2.15902] [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: 06/27/2023] [Revised: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024] Open
Abstract
Although zinc deficiency (secondary to malnutrition) has long been considered an important contributor to morbidity and mortality of infectious disease (e.g. diarrhea disorders), epidemiologic data (including randomized controlled trials with supplemental zinc) for such a role in lower respiratory tract infection are somewhat ambiguous. In the current study, we provide the first preclinical evidence demonstrating that although diet-induced acute zinc deficiency (Zn-D: ~50% decrease) did not worsen infection induced by either influenza A (H1N1) or methicillin-resistant staph aureus (MRSA), Zn-D mice were sensitive to the injurious effects of superinfection of H1N1 with MRSA. Although the mechanism underlying the sensitivity of ZnD mice to combined H1N1/MRSA infection is unclear, it was noteworthy that this combination exacerbated lung injury as shown by lung epithelial injury markers (increased BAL protein) and decreased genes related to epithelial integrity in Zn-D mice (surfactant protein C and secretoglobins family 1A member 1). As bacterial pneumonia accounts for 25%-50% of morbidity and mortality from influenza A infection, zinc deficiency may be an important pathology component of respiratory tract infections.
Collapse
Affiliation(s)
- Radha Gopal
- Department of PediatricsUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| | - Egemen Tutuncuoglu
- The Clinical Research, Investigation, and Systems Modeling of Acute Illness (CRISMA) CenterUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Present address:
Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Veli Bakalov
- The Clinical Research, Investigation, and Systems Modeling of Acute Illness (CRISMA) CenterUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Present address:
Medicine InstituteAllegheny Health NetworkPittsburghPennsylvaniaUSA
| | - Karla Wasserloos
- Department of Environmental and Occupational HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
- Present address:
R.D. 2PortersvillePennsylvaniaUSA
| | - HuiHua Li
- Department of Environmental and Occupational HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
- Present address:
Department of PathologyUniversity of WisconsinMadisonWisconsinUSA
| | - David Lemley
- Department of Environmental and Occupational HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
- Present address:
R.D. 2PortersvillePennsylvaniaUSA
| | - Louis J. DeVito
- Department of PediatricsUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| | | | - Douglas S. Reed
- Center for Vaccine ResearchUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Kevin J. McHugh
- Department of PediatricsUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| | - Baskaran Chinnappan
- Department of PediatricsUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| | - Alexis R. Andreas
- Department of Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Present address:
Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Abigail Maloy
- Department of Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Daniel Bain
- Department of Geology and Planetary ScienceUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - John F. Alcorn
- Department of PediatricsUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| | - Bruce R. Pitt
- Department of Environmental and Occupational HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Ata Murat Kaynar
- The Clinical Research, Investigation, and Systems Modeling of Acute Illness (CRISMA) CenterUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Anesthesiology and Perioperative MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
15
|
Mori A, Ohno H, Satoh-Takayama N. Disease pathogenesis and barrier functions regulated by group 3 innate lymphoid cells. Semin Immunopathol 2024; 45:509-519. [PMID: 38305897 DOI: 10.1007/s00281-024-01000-1] [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: 11/29/2023] [Accepted: 01/16/2024] [Indexed: 02/03/2024]
Abstract
The mucosal surface is in constant contact with foreign antigens and is regulated by unique mechanisms that are different from immune responses in the peripheral organs. For the last several decades, only adaptive immune cells such as helper T (Th) cells, Th1, Th2, or Th17 were targeted to study a wide variety of immune responses in the mucosal tissues. However, since their discovery, innate lymphoid cells (ILCs) have been attracting attention as a unique subset of immune cells that provide border defense with various functions and tissue specificity. ILCs are classified into different groups based on cell differentiation and functions. Group 3 innate lymphoid cells (ILC3s) are particularly in close proximity to mucosal surfaces and therefore have the opportunity to be exposed to a variety of bacteria including pathogenic bacteria. In recent years, studies have also provided much evidence that ILC3s contribute to disease pathogenesis as well as the defense of mucosal surfaces by rapidly responding to pathogens and coordinating other immune cells. As the counterpart of helper T cells, ILC3s together with other ILC subsets establish the immune balance between adaptive and innate immunity in protecting us from invasion or encounter with non-self-antigens for maintaining a complex homeostasis. In this review, we summarize recent advances in our understanding of ILCs, with a particular focus on the function of ILC3s in their involvement in bacterial infection and disease pathogenesis.
Collapse
Affiliation(s)
- Ayana Mori
- Immunobiology Laboratory, School of Science, Yokohama City University, 1-7-22, Suehiro, Tsurumi, Yokohama, 230-0045, Japan
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro, Tsurumi, Yokohama City, Kanagawa, 230-0045, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro, Tsurumi, Yokohama City, Kanagawa, 230-0045, Japan
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, 1-7-22, Suehiro, Tsurumi, Yokohama, 230-0045, Japan
- Laboratory for Immune Regulation, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Naoko Satoh-Takayama
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro, Tsurumi, Yokohama City, Kanagawa, 230-0045, Japan.
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, 1-7-22, Suehiro, Tsurumi, Yokohama, 230-0045, Japan.
| |
Collapse
|
16
|
Wang C, Khatun MS, Zhang Z, Allen MJ, Chen Z, Ellsworth CR, Currey JM, Dai G, Tian D, Bach K, Yin XM, Traina-Dorge V, Rappaport J, Maness NJ, Blair RV, Kolls JK, Pociask DA, Qin X. COVID-19 and influenza infections mediate distinct pulmonary cellular and transcriptomic changes. Commun Biol 2023; 6:1265. [PMID: 38092883 PMCID: PMC10719262 DOI: 10.1038/s42003-023-05626-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/22/2023] [Indexed: 12/17/2023] Open
Abstract
SARS-CoV-2 infection can cause persistent respiratory sequelae. However, the underlying mechanisms remain unclear. Here we report that sub-lethally infected K18-human ACE2 mice show patchy pneumonia associated with histiocytic inflammation and collagen deposition at 21 and 45 days post infection (DPI). Transcriptomic analyses revealed that compared to influenza-infected mice, SARS-CoV-2-infected mice had reduced interferon-gamma/alpha responses at 4 DPI and failed to induce keratin 5 (Krt5) at 6 DPI in lung, a marker of nascent pulmonary progenitor cells. Histologically, influenza- but not SARS-CoV-2-infected mice showed extensive Krt5+ "pods" structure co-stained with stem cell markers Trp63/NGFR proliferated in the pulmonary consolidation area at both 7 and 14 DPI, with regression at 21 DPI. These Krt5+ "pods" structures were not observed in the lungs of SARS-CoV-2-infected humans or nonhuman primates. These results suggest that SARS-CoV-2 infection fails to induce nascent Krt5+ cell proliferation in consolidated regions, leading to incomplete repair of the injured lung.
Collapse
Affiliation(s)
- Chenxiao Wang
- Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Mst Shamima Khatun
- Department of Medicine and Pediatrics, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Zhe Zhang
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Michaela J Allen
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Zheng Chen
- Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Calder R Ellsworth
- Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Joshua M Currey
- Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Guixiang Dai
- Department of Medicine and Pediatrics, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Di Tian
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Konrad Bach
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Xiao-Ming Yin
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Vicki Traina-Dorge
- Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Jay Rappaport
- Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Nicholas J Maness
- Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Robert V Blair
- Tulane National Primate Research Center, Covington, LA, 70433, USA
| | - Jay K Kolls
- Department of Medicine and Pediatrics, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Derek A Pociask
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| | - Xuebin Qin
- Tulane National Primate Research Center, Covington, LA, 70433, USA.
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
17
|
Moniruzzaman M, Rahman MA, Wang R, Wong KY, Chen ACH, Mueller A, Taylor S, Harding A, Illankoon T, Wiid P, Sajiir H, Schreiber V, Burr LD, McGuckin MA, Phipps S, Hasnain SZ. Interleukin-22 suppresses major histocompatibility complex II in mucosal epithelial cells. J Exp Med 2023; 220:e20230106. [PMID: 37695525 PMCID: PMC10494524 DOI: 10.1084/jem.20230106] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 05/22/2023] [Accepted: 08/18/2023] [Indexed: 09/12/2023] Open
Abstract
Major histocompatibility complex (MHC) II is dynamically expressed on mucosal epithelial cells and is induced in response to inflammation and parasitic infections, upon exposure to microbiota, and is increased in chronic inflammatory diseases. However, the regulation of epithelial cell-specific MHC II during homeostasis is yet to be explored. We discovered a novel role for IL-22 in suppressing epithelial cell MHC II partially via the regulation of endoplasmic reticulum (ER) stress, using animals lacking the interleukin-22-receptor (IL-22RA1), primary human and murine intestinal and respiratory organoids, and murine models of respiratory virus infection or with intestinal epithelial cell defects. IL-22 directly downregulated interferon-γ-induced MHC II on primary epithelial cells by modulating the expression of MHC II antigen A α (H2-Aα) and Class II transactivator (Ciita), a master regulator of MHC II gene expression. IL-22RA1-knockouts have significantly higher MHC II expression on mucosal epithelial cells. Thus, while IL-22-based therapeutics improve pathology in chronic disease, their use may increase susceptibility to viral infections.
Collapse
Affiliation(s)
- Md Moniruzzaman
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Immunopathology Group, Translational Research Institute, Mater Research Institute—The University of Queensland, Brisbane, Australia
| | - M. Arifur Rahman
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Immunopathology Group, Translational Research Institute, Mater Research Institute—The University of Queensland, Brisbane, Australia
| | - Ran Wang
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Immunopathology Group, Translational Research Institute, Mater Research Institute—The University of Queensland, Brisbane, Australia
| | - Kuan Yau Wong
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Immunopathology Group, Translational Research Institute, Mater Research Institute—The University of Queensland, Brisbane, Australia
| | - Alice C.-H. Chen
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Immunopathology Group, Translational Research Institute, Mater Research Institute—The University of Queensland, Brisbane, Australia
| | - Alexandra Mueller
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Immunopathology Group, Translational Research Institute, Mater Research Institute—The University of Queensland, Brisbane, Australia
| | - Steven Taylor
- South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Alexa Harding
- Immunopathology Group, Translational Research Institute, Mater Research Institute—The University of Queensland, Brisbane, Australia
| | - Thishan Illankoon
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Immunopathology Group, Translational Research Institute, Mater Research Institute—The University of Queensland, Brisbane, Australia
| | - Percival Wiid
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Immunopathology Group, Translational Research Institute, Mater Research Institute—The University of Queensland, Brisbane, Australia
| | - Haressh Sajiir
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Immunopathology Group, Translational Research Institute, Mater Research Institute—The University of Queensland, Brisbane, Australia
| | - Veronika Schreiber
- Immunopathology Group, Translational Research Institute, Mater Research Institute—The University of Queensland, Brisbane, Australia
| | - Lucy D. Burr
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Immunopathology Group, Translational Research Institute, Mater Research Institute—The University of Queensland, Brisbane, Australia
- Department of Respiratory and Sleep Medicine, Mater Health, South Brisbane, Australia
| | - Michael A. McGuckin
- Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Australia
| | - Simon Phipps
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Australia
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia
| | - Sumaira Z. Hasnain
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Immunopathology Group, Translational Research Institute, Mater Research Institute—The University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Australia
| |
Collapse
|
18
|
Al‐Kuraishy HM, Al‐Maiahy TJ, Al‐Gareeb AI, Alexiou A, Papadakis M, Elhussieny O, Saad HM, Batiha GE. New insights on the potential effect of progesterone in Covid-19: Anti-inflammatory and immunosuppressive effects. Immun Inflamm Dis 2023; 11:e1100. [PMID: 38018575 PMCID: PMC10683562 DOI: 10.1002/iid3.1100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 11/08/2023] [Accepted: 11/12/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is a pandemic disease caused by severe acute respiratory syndrome CoV type 2 (SARS-CoV-2). COVID-19 is higher in men than women and sex hormones have immune-modulator effects during different viral infections, including SARS-CoV-2 infection. One of the essential sex hormones is progesterone (P4). AIMS This review aimed to reveal the association between P4 and Covid-19. RESULTS AND DISCUSSION The possible role of P4 in COVID-19 could be beneficial through the modulation of inflammatory signaling pathways, induction of the release of anti-inflammatory cytokines, and inhibition release of pro-inflammatory cytokines. P4 stimulates skew of naïve T cells from inflammatory Th1 toward anti-inflammatory Th2 with activation release of anti-inflammatory cytokines, and activation of regulatory T cells (Treg) with decreased interferon-gamma production that increased during SARS-CoV-2 infection. In addition, P4 is regarded as a potent antagonist of mineralocorticoid receptor (MR), it could reduce MRs that were activated by stimulated aldosterone from high AngII during SARS-CoV-2. P4 active metabolite allopregnanolone is regarded as a neurosteroid that acts as a positive modulator of γ-aminobutyric acid (GABAA ) so it may reduce neuropsychiatric manifestations and dysautonomia in COVID-19 patients. CONCLUSION Taken together, the anti-inflammatory and immunomodulatory properties of P4 may improve central and peripheral complications in COVID-19.
Collapse
Affiliation(s)
- Hayder M. Al‐Kuraishy
- Department of Clinical Pharmacology and Therapeutic Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | - Thabat J. Al‐Maiahy
- Department of Gynecology and Obstetrics, College of MedicineAl‐Mustansiriyah UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Therapeutic Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | - Athanasios Alexiou
- University Centre for Research & DevelopmentChandigarh UniversityMohaliPunjabIndia
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
- Department of Research & DevelopmentAFNP MedWienAustria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten‐Herdecke, Heusnerstrasse 40University of Witten‐HerdeckeWuppertalGermany
| | - Omnya Elhussieny
- Department of Histology and Cytology, Faculty of Veterinary MedicineMatrouh UniversityMarsa MatruhEgypt
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary MedicineMatrouh UniversityMarsa MatruhEgypt
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour University, DamanhourAlBeheiraEgypt
| |
Collapse
|
19
|
Narasimhan M, Muthukumar A, Sataranatarajan K, Mahimainathan L, Mahan L, Timofte I, Bollineni S, Joerns J, Zhang S, Gorman A, Banga A, Mohanka M, Torres F, Lawrence A, Thalachallour M, Kaza V. Crossroads between Autoimmunity and COVID-19 in Lung Transplant Recipients. Viruses 2023; 15:2045. [PMID: 37896822 PMCID: PMC10612071 DOI: 10.3390/v15102045] [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: 08/22/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
The presence of a certain group of auto-antibodies (AAbs) is known to correlate with the severity of COVID-19. It is, however, unknown if such AAbs are prevalent and impact COVID-19-related outcomes in lung transplant recipients (LTRs) who are immunosuppressed. We performed a retrospective study of LTRs with COVID-19 and analyzed samples before and after COVID-19 for IgG AAbs. AAbs analysis was carried out using autoimmune and coronavirus microarray and the resulting cross-sectional differences in Ab-scores and clinical variables were analyzed using Fischer's Exact test for categorical variables and a paired t-test for continuous variables. Linear regression was used to analyze the differences in Ab-scores and COVID-19 severity. LTRs with non-severe [NS gp (n = 10)], and severe [S gp (n = 8)] COVID-19 disease were included. Ferritin and acute respiratory failure were higher in the S group (p = 0.03; p < 0.0001). Among the AAbs analyzed, interferon-related AAbs (IFN-alpha2, IFN-beta, IFN lamba, IFN-epsilon), eight interleukin-related AAbs, and several tissue-related AAbs were also found to be changed significantly from pre- to post-COVID-19 (p < 0.05). IFN-lambda (p = 0.03) and IL-22 (p = 0.002) were significantly associated with COVID-19 severity and remained significant in linear regression analysis while controlling for other variables. AAbs are common in LTRs, and certain groups of antibodies are particularly enhanced in LTRs with severe COVID-19. Preliminary observations of this study need to be confirmed by a larger sample size.
Collapse
Affiliation(s)
- Madhusudhanan Narasimhan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (M.N.); (A.M.); (K.S.); (L.M.)
| | - Alagarraju Muthukumar
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (M.N.); (A.M.); (K.S.); (L.M.)
| | - Kavithalakshmi Sataranatarajan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (M.N.); (A.M.); (K.S.); (L.M.)
| | - Lenin Mahimainathan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (M.N.); (A.M.); (K.S.); (L.M.)
| | - Luke Mahan
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.M.); (I.T.); (S.B.); (J.J.); (A.B.); (M.M.); (F.T.); (A.L.)
| | - Irina Timofte
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.M.); (I.T.); (S.B.); (J.J.); (A.B.); (M.M.); (F.T.); (A.L.)
| | - Srinivas Bollineni
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.M.); (I.T.); (S.B.); (J.J.); (A.B.); (M.M.); (F.T.); (A.L.)
| | - John Joerns
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.M.); (I.T.); (S.B.); (J.J.); (A.B.); (M.M.); (F.T.); (A.L.)
| | - Song Zhang
- Peter O’Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (S.Z.); (A.G.)
| | - April Gorman
- Peter O’Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (S.Z.); (A.G.)
| | - Amit Banga
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.M.); (I.T.); (S.B.); (J.J.); (A.B.); (M.M.); (F.T.); (A.L.)
| | - Manish Mohanka
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.M.); (I.T.); (S.B.); (J.J.); (A.B.); (M.M.); (F.T.); (A.L.)
| | - Fernando Torres
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.M.); (I.T.); (S.B.); (J.J.); (A.B.); (M.M.); (F.T.); (A.L.)
| | - Adrian Lawrence
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.M.); (I.T.); (S.B.); (J.J.); (A.B.); (M.M.); (F.T.); (A.L.)
| | | | - Vaidehi Kaza
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.M.); (I.T.); (S.B.); (J.J.); (A.B.); (M.M.); (F.T.); (A.L.)
| |
Collapse
|
20
|
Beppu AK, Zhao J, Yao C, Carraro G, Israely E, Coelho AL, Drake K, Hogaboam CM, Parks WC, Kolls JK, Stripp BR. Epithelial plasticity and innate immune activation promote lung tissue remodeling following respiratory viral infection. Nat Commun 2023; 14:5814. [PMID: 37726288 PMCID: PMC10509177 DOI: 10.1038/s41467-023-41387-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 09/02/2023] [Indexed: 09/21/2023] Open
Abstract
Epithelial plasticity has been suggested in lungs of mice following genetic depletion of stem cells but is of unknown physiological relevance. Viral infection and chronic lung disease share similar pathological features of stem cell loss in alveoli, basal cell (BC) hyperplasia in small airways, and innate immune activation, that contribute to epithelial remodeling and loss of lung function. We show that a subset of distal airway secretory cells, intralobar serous (IS) cells, are activated to assume BC fates following influenza virus infection. Injury-induced hyperplastic BC (hBC) differ from pre-existing BC by high expression of IL-22Ra1 and undergo IL-22-dependent expansion for colonization of injured alveoli. Resolution of virus-elicited inflammation results in BC to IS re-differentiation in repopulated alveoli, and increased local expression of protective antimicrobial factors, but fails to restore normal alveolar epithelium responsible for gas exchange.
Collapse
Affiliation(s)
- Andrew K Beppu
- Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Department of Medicine, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Juanjuan Zhao
- Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Department of Medicine, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Changfu Yao
- Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Department of Medicine, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Gianni Carraro
- Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Department of Medicine, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Edo Israely
- Department of Medicine, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Anna Lucia Coelho
- Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Katherine Drake
- Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Department of Medicine, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Cory M Hogaboam
- Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - William C Parks
- Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Jay K Kolls
- Tulane Center for Translational Research in Infection and Inflammation, School of Medicine, New Orleans, LA, 70112, USA
| | - Barry R Stripp
- Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
- Department of Medicine, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
| |
Collapse
|
21
|
Motta F, Tonutti A, Isailovic N, Ceribelli A, Costanzo G, Rodolfi S, Selmi C, De Santis M. Proteomic aptamer analysis reveals serum biomarkers associated with disease mechanisms and phenotypes of systemic sclerosis. Front Immunol 2023; 14:1246777. [PMID: 37753072 PMCID: PMC10518467 DOI: 10.3389/fimmu.2023.1246777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
Background Systemic sclerosis (SSc) is an autoimmune connective tissue disease that affects multiple organs, leading to elevated morbidity and mortality with limited treatment options. The early detection of organ involvement is challenging as there is currently no serum marker available to predict the progression of SSc. The aptamer technology proteomic analysis holds the potential to correlate SSc manifestations with serum proteins up to femtomolar concentrations. Methods This is a two-tier study of serum samples from women with SSc (including patients with interstitial lung disease - ILD - at high-resolution CT scan) and age-matched healthy controls (HC) that were first analyzed with aptamer-based proteomic analysis for over 1300 proteins. Proposed associated proteins were validated by ELISA first in an independent cohort of patients with SSc and HC, and selected proteins subject to further validation in two additional cohorts. Results The preliminary aptamer-based proteomic analysis identified 33 proteins with significantly different concentrations in SSc compared to HC sera and 9 associated with SSc-ILD, including proteins involved in extracellular matrix formation and cell-cell adhesion, angiogenesis, leukocyte recruitment, activation, and signaling. Further validations in independent cohorts ultimately confirmed the association of specific proteins with early SSc onset, specific organ involvement, and serum autoantibodies. Conclusions Our multi-tier proteomic analysis identified serum proteins discriminating patients with SSc and HC or associated with different SSc subsets, disease duration, and manifestations, including ILD, skin involvement, esophageal disease, and autoantibodies.
Collapse
Affiliation(s)
- Francesca Motta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Rheumatology and Clinical Immunology, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
| | - Antonio Tonutti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Rheumatology and Clinical Immunology, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
| | - Natasa Isailovic
- Rheumatology and Clinical Immunology, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
| | - Angela Ceribelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Rheumatology and Clinical Immunology, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
| | - Giovanni Costanzo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Rheumatology and Clinical Immunology, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
| | - Stefano Rodolfi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Rheumatology and Clinical Immunology, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
| | - Carlo Selmi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Rheumatology and Clinical Immunology, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
| | - Maria De Santis
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Rheumatology and Clinical Immunology, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy
| |
Collapse
|
22
|
Seth P, Dubey S. IL-22 as a target for therapeutic intervention: Current knowledge on its role in various diseases. Cytokine 2023; 169:156293. [PMID: 37441942 DOI: 10.1016/j.cyto.2023.156293] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/12/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023]
Abstract
IL-22 has emerged as a crucial cytokine mediating protective response against pathogens and tissue regeneration. Dysregulated production of IL-22 has been shown to play a pivotal role in the pathogenesis of various diseases like malignant tumours, viral, cardiovascular, allergic and autoimmune disorders. Interleukin 22 belongs to IFN-IL-10 cytokine family. It is a major proinflammatory cytokine secreted by activated Th1 cells (Th22), though can also be secreted by many other immune cells like group 3 innate lymphocytes, γδ T cells, NK cells, NK T cells, and mucosal associated invariant T cells. Th22 cells exclusively release IL-22 but not IL-17 or IFN-γ (as Th1 cells releases IFN-γ along with IL-22 and Th17 cells releases IL-17 along with IL-22) and also express aryl hydrocarbon receptor as the key transcription factor. Th22 cells also exhibit expression of chemokine receptor CCR6 and skin-homing receptors CCR4 and CCR10 indicating the involvement of this subset in bolstering epithelial barrier immunity and promoting secretion of antimicrobial peptides (AMPs) from intestinal epithelial cells. The function of IL-22 is modulated by IL-22 binding protein (binds to IL-22 and inhibits it binding to its cell surface receptor); which serves as a competitor for IL-22R1 chain of IL-22 receptor. The pathogenic and protective nature of the Th22 cells is modulated both by the site of infected tissue and the type of disease pathology. This review aims to discuss key features of IL-22 biology, comparisons between IL and 22 and IFN-γ and its role as a potential immune therapy target in different maladies.
Collapse
Affiliation(s)
- Pranav Seth
- Amity Institute of Virology & Immunology, Amity University Uttar Pradesh, Sector 125, Noida, India
| | - Shweta Dubey
- Amity Institute of Virology & Immunology, Amity University Uttar Pradesh, Sector 125, Noida, India.
| |
Collapse
|
23
|
Liu L, Liu R, Wei C, Li D, Gao X. The role of IL-17 in lung cancer growth. Cytokine 2023; 169:156265. [PMID: 37348188 DOI: 10.1016/j.cyto.2023.156265] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/27/2023] [Accepted: 06/01/2023] [Indexed: 06/24/2023]
Abstract
Interleukin 17 (IL-17) is an inflammatory cytokine with multiple roles in immune protection, immunopathology, and inflammation-related tumors. Lung cancer is inflammation-related cancer, and a large number of studies have shown that IL-17 contributes to the metastasis and progression of lung cancer. However, some studies have shown that IL17 inhibits the occurrence of lung cancer. At present, there is still some controversy about the role of IL17 in the occurrence and development of lung cancer. This review introduces the basic characteristics of IL-17 and focuses on its role in lung cancer, in order to provide a certain theoretical basis for the prevention, diagnosis, and treatment of lung cancer.
Collapse
Affiliation(s)
- Liping Liu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Renli Liu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Chaojie Wei
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Dong Li
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China.
| | - Xiuzhu Gao
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, China.
| |
Collapse
|
24
|
Holloman BL, Cannon A, Wilson K, Nagarkatti P, Nagarkatti M. Aryl Hydrocarbon Receptor Activation Ameliorates Acute Respiratory Distress Syndrome through Regulation of Th17 and Th22 Cells in the Lungs. mBio 2023; 14:e0313722. [PMID: 36809070 PMCID: PMC10128024 DOI: 10.1128/mbio.03137-22] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 02/23/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is triggered by a variety of insults, including bacterial and viral infections, and this leads to high mortality. While the role of the aryl hydrocarbon receptor (AhR) in mucosal immunity is being increasingly recognized, its function during ARDS is unclear. In the current study, we investigated the role of AhR in LPS-induced ARDS. AhR ligand, indole-3-carbinol (I3C), attenuated ARDS which was associated with a decrease in CD4+ RORγt +IL-17a+IL-22+ pathogenic Th17 cells, but not CD4+RORγt +IL-17a+IL-22- homeostatic Th 17 cells, in the lungs. AhR activation also led to a significant increase in CD4+IL-17a-IL-22+ Th22 cells. I3C-mediated Th22 cell expansion was dependent on the AhR expression on RORγt+ cells. AhR activation downregulated miR-29b-2-5p in immune cells from the lungs, which in turn downregulated RORc expression and upregulated IL-22. Collectively, the current study suggests that AhR activation can attenuate ARDS and may serve as a therapeutic modality by which to treat this complex disorder. IMPORTANCE Acute respiratory distress syndrome (ARDS) is a type of respiratory failure that is triggered by a variety of bacterial and viral infections, including the coronavirus SARS-CoV2. ARDS is associated with a hyperimmune response in the lungs that which is challenging to treat. Because of this difficulty, approximately 40% of patients with ARDS die. Thus, it is critical to understand the nature of the immune response that is functional in the lungs during ARDS as well as approaches by which to attenuate it. AhR is a transcription factor that is activated by a variety of endogenous and exogenous environmental chemicals as well as bacterial metabolites. While AhR has been shown to regulate inflammation, its role in ARDS is unclear. In the current study, we provide evidence that AhR activation can attenuate LPS-mediated ARDS through the activation of Th22 cells in the lungs, which are regulated through miR-29b-2-5p. Thus, AhR can be targeted to attenuate ARDS.
Collapse
Affiliation(s)
- Bryan Latrell Holloman
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Alkeiver Cannon
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Kiesha Wilson
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| |
Collapse
|
25
|
Shi C, Su C, Cen L, Han L, Tang J, Wang Z, Shi X, Ju D, Cao Y, Zhu H. Vunakizumab-IL22, a Novel Fusion Protein, Promotes Intestinal Epithelial Repair and Protects against Gut Injury Induced by the Influenza Virus. Biomedicines 2023; 11:biomedicines11041160. [PMID: 37189778 DOI: 10.3390/biomedicines11041160] [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: 03/03/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
Secondary immune damage to the intestinal mucosa due to an influenza virus infection has gained the attention of investigators. The protection of the intestinal barrier is an effective means of improving the survival rate in cases of severe pneumonia. We developed a fusion protein, Vunakizumab-IL22(vmab-IL22), by combining an anti-IL17A antibody with IL22. Our previous study showed that Vunakizumab-IL22 repairs the pulmonary epithelial barrier in influenza virus-infected mice. In this study, we investigated the protective effects against enteritis given its anti-inflammatory and tissue repair functions. The number of goblet cells and the expression of zonula occludens protein 1(ZO-1), Mucin-2, Ki67 and IL-22R were determined by immunohistochemistry (IHC) and quantitative RT-PCR in influenza A virus (H1N1)-infected mice. The expression of NOD-like receptor pyrin domain containing 3 (NLRP3) and toll- like-receptor-4 (TLR4) was assayed by IHC in the lungs and intestine in HIN1 virus-induced mice to evaluate the whole efficacy of the protective effects on lungs and intestines. Consequently, Cytochrome C, phosphorylation of nuclear factor NF-kappaB (p-NF-κB), IL-1β, NLRP3 and Caspase 3 were assayed by Western blotting in dextran sulfate sodium salt (DSS)-treated mice. Treatment with Vunakizumab-IL22 improved the shortened colon length, macroscopic and microscopic morphology of the small intestine (p < 0.001) significantly, and strengthened the tight junction proteins, which was accompanied with the upregulated expression of IL22R. Meanwhile, Vunakizumab-mIL22 inhibited the expression of inflammation-related protein in a mouse model of enteritis induced by H1N1 and DSS. These findings provide new evidence for the treatment strategy for severe viral pneumonia involved in gut barrier protection. The results suggest that Vunakizumab-IL22 is a promising biopharmaceutical drug and is a candidate for the treatment of direct and indirect intestinal injuries, including those induced by the influenza virus and DSS.
Collapse
Affiliation(s)
- Chenchen Shi
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunoTherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Division of Spine, Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Chang Su
- Department of Surgery, Minhang Hospital, Fudan University, Shanghai 201100, China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201100, China
| | - Lifeng Cen
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunoTherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Lei Han
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunoTherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jianguo Tang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Zetian Wang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Xunlong Shi
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunoTherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Dianwen Ju
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunoTherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yiou Cao
- Department of Surgery, Minhang Hospital, Fudan University, Shanghai 201100, China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201100, China
| | - Haiyan Zhu
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunoTherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
26
|
Resnick JD, Beer MA, Pekosz A. Early Transcriptional Responses of Human Nasal Epithelial Cells to Infection with Influenza A and SARS-CoV-2 Virus Differ and Are Influenced by Physiological Temperature. Pathogens 2023; 12:480. [PMID: 36986402 PMCID: PMC10051809 DOI: 10.3390/pathogens12030480] [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: 12/30/2022] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Influenza A (IAV) and SARS-CoV-2 (SCV2) viruses represent an ongoing threat to public health. Both viruses target the respiratory tract, which consists of a gradient of cell types, receptor expression, and temperature. Environmental temperature has been an understudied contributor to infection susceptibility and understanding its impact on host responses to infection could help uncover new insight into severe disease risk factors. As the nasal passageways are the initial site of respiratory virus infection, in this study we investigated the effect of temperature on host responses in human nasal epithelial cells (hNECs) utilizing IAV and SCV2 in vitro infection models. We demonstrate that temperature affected SCV2, but not IAV, viral replicative fitness and that SCV2-infected cultures were slower to mount an infection-induced response, likely due to suppression by the virus. Additionally, we show that that temperature not only changed the basal transcriptomic landscape of epithelial cells, but that it also impacted the response to infection. The induction of interferon and other innate immune responses was not drastically affected by temperature, suggesting that while the baseline antiviral response at different temperatures remained consistent, there may be metabolic or signaling changes that affect how well the cultures were able to adapt to new pressures, such as infection. Finally, we show that hNECs responded differently to IAV and SCV2 infection in ways that give insight into how the virus is able to manipulate the cell to allow for replication and release. Taken together, these data give new insight into the innate immune response to respiratory infections and can assist in identifying new treatment strategies for respiratory infections.
Collapse
Affiliation(s)
- Jessica D. Resnick
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael A. Beer
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
27
|
Resnick JD, Beer MA, Pekosz A. Early transcriptional responses of human nasal epithelial cells to infection with Influenza A and SARS-CoV-2 virus differ and are influenced by physiological temperature. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531609. [PMID: 36945583 PMCID: PMC10028862 DOI: 10.1101/2023.03.07.531609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
Influenza A (IAV) and SARS-CoV-2 (SCV2) viruses represent an ongoing threat to public health. Both viruses target the respiratory tract, which consists of a gradient of cell types, receptor expression, and temperature. Environmental temperature has been an un-derstudied contributor to infection susceptibility and understanding its impact on host responses to infection could help uncover new insights into severe disease risk factors. As the nasal passageways are the initial site of respiratory virus infection, in this study we investigated the effect of temperature on host responses in human nasal epithelial cells (hNECs) utilizing IAV and SCV2 in vitro infection models. We demonstrate that temperature affects SCV2, but not IAV, viral replicative fitness and that SCV2 infected cultures are slower to mount an infection-induced response, likely due to suppression by the virus. Additionally, we show that that temperature not only changes the basal transcriptomic landscape of epithelial cells, but that it also impacts the response to infection. The induction of interferon and other innate immune responses were not drastically affected by temperature, suggesting that while the baseline antiviral response at different temperatures remains consistent, there may be metabolic or signaling changes that affect how well the cultures are able to adapt to new pressures such as infection. Finally, we show that hNECs respond differently to IAV and SCV2 infection in ways that give insight into how the virus is able to manipulate the cell to allow for replication and release. Taken together, these data give new insight into the innate immune response to respiratory infections and can assist in identifying new treatment strategies for respiratory infections.
Collapse
Affiliation(s)
- Jessica D Resnick
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- McKusick- Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael A Beer
- McKusick- Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
28
|
Zhang K, Chen L, Zhu C, Zhang M, Liang C. Current Knowledge of Th22 Cell and IL-22 Functions in Infectious Diseases. Pathogens 2023; 12:pathogens12020176. [PMID: 36839448 PMCID: PMC9965464 DOI: 10.3390/pathogens12020176] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/19/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
T helper 22 (Th22) cells, a newly defined CD4+ T-cell lineage, are characterized by their distinct cytokine profile, which primarily consists of IL-13, IL-22 and TNF-α. Th22 cells express a wide spectrum of chemokine receptors, such as CCR4, CCR6 and CCR10. The main effector molecule secreted by Th22 cells is IL-22, a member of the IL-10 family, which acts by binding to IL-22R and triggering a complex downstream signaling system. Th22 cells and IL-22 have been found to play variable roles in human immunity. In preventing the progression of infections such as HIV and influenza, Th22/IL-22 exhibited protective anti-inflammatory characteristics, and their deleterious proinflammatory activities have been demonstrated to exacerbate other illnesses, including hepatitis B and Helicobacter pylori infection. Herein, we review the current understanding of Th22 cells, including their definition, differentiation and mechanisms, and the effect of Th22/IL-22 on human infectious diseases. According to studies on Th22 cells, Th22/IL-22 may be a promising therapeutic target and an effective treatment strategy for various infections.
Collapse
Affiliation(s)
- Kunyu Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230022, China
- Institute of Urology, Anhui Medical University, Hefei 230022, China
- The Second Clinical Medical College, Anhui Medical University, Hefei 230032, China
| | - Lei Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230022, China
- Institute of Urology, Anhui Medical University, Hefei 230022, China
| | - Chenyu Zhu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230022, China
- Institute of Urology, Anhui Medical University, Hefei 230022, China
- The Second Clinical Medical College, Anhui Medical University, Hefei 230032, China
| | - Meng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230022, China
- Institute of Urology, Anhui Medical University, Hefei 230022, China
- Correspondence: (M.Z.); (C.L.); Tel./Fax: +86-55162922034 (M.Z.); +86-55162922034 (C.L.)
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230022, China
- Institute of Urology, Anhui Medical University, Hefei 230022, China
- Correspondence: (M.Z.); (C.L.); Tel./Fax: +86-55162922034 (M.Z.); +86-55162922034 (C.L.)
| |
Collapse
|
29
|
Tao H, Xu Y, Zhang S. The Role of Macrophages and Alveolar Epithelial Cells in the Development of ARDS. Inflammation 2023; 46:47-55. [PMID: 36048270 PMCID: PMC9435414 DOI: 10.1007/s10753-022-01726-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/25/2022] [Accepted: 08/01/2022] [Indexed: 11/26/2022]
Abstract
Acute lung injury (ALI) usually causes acute respiratory distress syndrome (ARDS), or even death in critical ill patients. Immune cell infiltration in inflamed lungs is an important hallmark of ARDS. Macrophages are a type of immune cell that participate in the entire pathogenic trajectory of ARDS and most prominently via their interactions with lung alveolar epithelial cells (AECs). In the early stage of ARDS, classically activated macrophages secrete pro-inflammatory cytokines to clearance of the pathogens which may damage alveolar AECs cell structure and result in cell death. Paradoxically, in late stage of ARDS, anti-inflammatory cytokines secreted by alternatively activated macrophages dampen the inflammation response and promote epithelial regeneration and alveolar structure remodeling. In this review, we discuss the important role of macrophages and AECs in the progression of ARDS.
Collapse
Affiliation(s)
- Huan Tao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430033, China
| | - Younian Xu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430033, China.
| | - Shihai Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430033, China.
| |
Collapse
|
30
|
Astegolimab or Efmarodocokin Alfa in Patients With Severe COVID-19 Pneumonia: A Randomized, Phase 2 Trial. Crit Care Med 2023; 51:103-116. [PMID: 36519984 PMCID: PMC9749945 DOI: 10.1097/ccm.0000000000005716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Severe cases of COVID-19 pneumonia can lead to acute respiratory distress syndrome (ARDS). Release of interleukin (IL)-33, an epithelial-derived alarmin, and IL-33/ST2 pathway activation are linked with ARDS development in other viral infections. IL-22, a cytokine that modulates innate immunity through multiple regenerative and protective mechanisms in lung epithelial cells, is reduced in patients with ARDS. This study aimed to evaluate safety and efficacy of astegolimab, a human immunoglobulin G2 monoclonal antibody that selectively inhibits the IL-33 receptor, ST2, or efmarodocokin alfa, a human IL-22 fusion protein that activates IL-22 signaling, for treatment of severe COVID-19 pneumonia. DESIGN Phase 2, double-blind, placebo-controlled study (COVID-astegolimab-IL). SETTING Hospitals. PATIENTS Hospitalized adults with severe COVID-19 pneumonia. INTERVENTIONS Patients were randomized to receive IV astegolimab, efmarodocokin alfa, or placebo, plus standard of care. The primary endpoint was time to recovery, defined as time to a score of 1 or 2 on a 7-category ordinal scale by day 28. MEASUREMENTS AND MAIN RESULTS The study randomized 396 patients. Median time to recovery was 11 days (hazard ratio [HR], 1.01 d; p = 0.93) and 10 days (HR, 1.15 d; p = 0.38) for astegolimab and efmarodocokin alfa, respectively, versus 10 days for placebo. Key secondary endpoints (improved recovery, mortality, or prevention of worsening) showed no treatment benefits. No new safety signals were observed and adverse events were similar across treatment arms. Biomarkers demonstrated that both drugs were pharmacologically active. CONCLUSIONS Treatment with astegolimab or efmarodocokin alfa did not improve time to recovery in patients with severe COVID-19 pneumonia.
Collapse
|
31
|
Sullivan KM, Matthay MA. Lessons From a Negative Clinical Trial: Novel Immunological Targets for COVID-19 and Beyond. Crit Care Med 2023; 51:153-156. [PMID: 36519993 PMCID: PMC9749943 DOI: 10.1097/ccm.0000000000005719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Kathryn M Sullivan
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Michael A Matthay
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA
- Cardiovascular Research Institute, Departments of Medicine and Anesthesia, University of California San Francisco, San Francisco, CA
| |
Collapse
|
32
|
Esnault S, Jarjour NN. Development of Adaptive Immunity and Its Role in Lung Remodeling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1426:287-351. [PMID: 37464127 DOI: 10.1007/978-3-031-32259-4_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Asthma is characterized by airflow limitations resulting from bronchial closure, which can be either reversible or fixed due to changes in airway tissue composition and structure, also known as remodeling. Airway remodeling is defined as increased presence of mucins-producing epithelial cells, increased thickness of airway smooth muscle cells, angiogenesis, increased number and activation state of fibroblasts, and extracellular matrix (ECM) deposition. Airway inflammation is believed to be the main cause of the development of airway remodeling in asthma. In this chapter, we will review the development of the adaptive immune response and the impact of its mediators and cells on the elements defining airway remodeling in asthma.
Collapse
|
33
|
Biryukov SS, Cote CK, Klimko CP, Dankmeyer JL, Rill NO, Shoe JL, Hunter M, Shamsuddin Z, Velez I, Hedrick ZM, Rosario-Acevedo R, Talyansky Y, Schmidt LK, Orne CE, Fetterer DP, Burtnick MN, Brett PJ, Welkos SL, DeShazer D. Evaluation of two different vaccine platforms for immunization against melioidosis and glanders. Front Microbiol 2022; 13:965518. [PMID: 36060742 PMCID: PMC9428723 DOI: 10.3389/fmicb.2022.965518] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/22/2022] [Indexed: 11/13/2022] Open
Abstract
Burkholderia pseudomallei and the closely related species, Burkholderia mallei, produce similar multifaceted diseases which range from rapidly fatal to protracted and chronic, and are a major cause of mortality in endemic regions. Besides causing natural infections, both microbes are Tier 1 potential biothreat agents. Antibiotic treatment is prolonged with variable results, hence effective vaccines are urgently needed. The purpose of our studies was to compare candidate vaccines that target both melioidosis and glanders to identify the most efficacious one(s) and define residual requirements for their transition to the non-human primate aerosol model. Studies were conducted in the C57BL/6 mouse model to evaluate the humoral and cell-mediated immune response and protective efficacy of three Burkholderia vaccine candidates against lethal aerosol challenges with B. pseudomallei K96243, B. pseudomallei MSHR5855, and B. mallei FMH. The recombinant vaccines generated significant immune responses to the vaccine antigens, and the live attenuated vaccine generated a greater immune response to OPS and the whole bacterial cells. Regardless of the candidate vaccine evaluated, the protection of mice was associated with a dampened cytokine response within the lungs after exposure to aerosolized bacteria. Despite being delivered by two different platforms and generating distinct immune responses, two experimental vaccines, a capsule conjugate + Hcp1 subunit vaccine and the live B. pseudomallei 668 ΔilvI strain, provided significant protection and were down-selected for further investigation and advanced development.
Collapse
Affiliation(s)
- Sergei S. Biryukov
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Christopher K. Cote
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
- *Correspondence: Christopher K. Cote
| | - Christopher P. Klimko
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Jennifer L. Dankmeyer
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Nathaniel O. Rill
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Jennifer L. Shoe
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Melissa Hunter
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Zain Shamsuddin
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Ivan Velez
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Zander M. Hedrick
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Raysa Rosario-Acevedo
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Yuli Talyansky
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Lindsey K. Schmidt
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, United States
| | - Caitlyn E. Orne
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, United States
| | - David P. Fetterer
- Biostatistics Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - Mary N. Burtnick
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, United States
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Paul J. Brett
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, United States
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Susan L. Welkos
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
| | - David DeShazer
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, United States
- David DeShazer
| |
Collapse
|
34
|
Fang S, Ju D, Lin Y, Chen W. The role of interleukin-22 in lung health and its therapeutic potential for COVID-19. Front Immunol 2022; 13:951107. [PMID: 35967401 PMCID: PMC9364265 DOI: 10.3389/fimmu.2022.951107] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Although numerous clinical trials have been implemented, an absolutely effective treatment against coronavirus disease 2019 (COVID-19) is still elusive. Interleukin-22 (IL-22) has attracted great interest over recent years, making it one of the best-studied cytokines of the interleukin-10 (IL-10) family. Unlike most interleukins, the major impact of IL-22 is exclusively on fibroblasts and epithelial cells due to the restricted expression of receptor. Numerous studies have suggested that IL-22 plays a crucial role in anti-viral infections through significantly ameliorating the immune cell-mediated inflammatory responses, and reducing tissue injury as well as further promoting epithelial repair and regeneration. Herein, we pay special attention to the role of IL-22 in the lungs. We summarize the latest progress in our understanding of IL-22 in lung health and disease and further discuss maneuvering this cytokine as potential immunotherapeutic strategy for the effective manage of COVID-19.
Collapse
Affiliation(s)
- Si Fang
- Multiscale Research Institute of Complex Systems & Jingan District Central Hospital of Shanghai, Fudan University, Shanghai, China
| | - Dianwen Ju
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai, China
| | - Yong Lin
- Multiscale Research Institute of Complex Systems & Jingan District Central Hospital of Shanghai, Fudan University, Shanghai, China
| | - Wei Chen
- Multiscale Research Institute of Complex Systems & Jingan District Central Hospital of Shanghai, Fudan University, Shanghai, China
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, United States
| |
Collapse
|
35
|
Ulu A, Sveiven S, Bilg A, Velazquez JV, Diaz M, Mukherjee M, Yuil-Valdes AG, Kota S, Burr A, Najera A, Nordgren TM. IL-22 regulates inflammatory responses to agricultural dust-induced airway inflammation. Toxicol Appl Pharmacol 2022; 446:116044. [PMID: 35525330 PMCID: PMC9133182 DOI: 10.1016/j.taap.2022.116044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/21/2022] [Accepted: 04/28/2022] [Indexed: 11/26/2022]
Abstract
IL-22 is a unique cytokine that is upregulated in many chronic inflammatory diseases, including asthma, and modulates tissue responses during inflammation. However, the role of IL-22 in the resolution of inflammation and how this contributes to lung repair processes are largely unknown. Here, we tested the hypothesis that IL-22 signaling is critical in inflammation resolution after repetitive exposure to agricultural dust. Using an established mouse model of organic dust extract-induced lung inflammation, we found that IL-22 knockout mice have an enhanced response to agricultural dust as evidenced by an exacerbated increase in infiltrating immune cells and lung pathology as compared to wild-type controls. We further identified that, in response to dust, IL-22 is expressed in airway epithelium and in Ym1+ macrophages found within the parenchyma in response to dust. The increase in IL-22 expression was accompanied by increases in IL-22 receptor IL-22R1 within the lung epithelium. In addition, we found that alveolar macrophages in vivo as well as THP-1 cells in vitro express IL-22, and this expression is modulated by dust exposure. Furthermore, subcellular localization of IL-22 appears to be in the Golgi of resting THP1 human monocytes, and treatment with dust extracts is associated with IL-22 release into the cytosolic compartment from the Golgi reservoirs during dust extract exposure. Taken together, we have identified a significant role for macrophage-mediated IL-22 signaling that is activated in dust-induced lung inflammation in mice.
Collapse
Affiliation(s)
- Arzu Ulu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Stefanie Sveiven
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Amanpreet Bilg
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Jalene V Velazquez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Marissa Diaz
- Riverside Community College, Riverside, CA 92521, USA
| | - Maheswari Mukherjee
- Department of Medical Sciences, College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ana G Yuil-Valdes
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Santosh Kota
- Department of Preprofessional Biology, University of Florida, Gainesville, FL 32603, USA
| | - Abigail Burr
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Aileen Najera
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Tara M Nordgren
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA; Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, 80521, USA.
| |
Collapse
|
36
|
Downey J, Randolph HE, Pernet E, Tran KA, Khader SA, King IL, Barreiro LB, Divangahi M. Mitochondrial cyclophilin D promotes disease tolerance by licensing NK cell development and IL-22 production against influenza virus. Cell Rep 2022; 39:110974. [PMID: 35732121 DOI: 10.1016/j.celrep.2022.110974] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 03/25/2022] [Accepted: 05/26/2022] [Indexed: 11/03/2022] Open
Abstract
Severity of pulmonary viral infections, including influenza A virus (IAV), is linked to excessive immunopathology, which impairs lung function. Thus, the same immune responses that limit viral replication can concomitantly cause lung damage that must be countered by largely uncharacterized disease tolerance mechanisms. Here, we show that mitochondrial cyclophilin D (CypD) protects against IAV via disease tolerance. CypD-/- mice are significantly more susceptible to IAV infection despite comparable antiviral immunity. This susceptibility results from damage to the lung epithelial barrier caused by a reduction in interleukin-22 (IL-22)-producing natural killer (NK) cells. Transcriptomic and functional data reveal that CypD-/- NK cells are immature and have altered cellular metabolism and impaired IL-22 production, correlating with dysregulated bone marrow lymphopoiesis. Administration of recombinant IL-22 or transfer of wild-type (WT) NK cells abrogates pulmonary damage and protects CypD-/- mice after IAV infection. Collectively, these results demonstrate a key role for CypD in NK cell-mediated disease tolerance.
Collapse
Affiliation(s)
- Jeffrey Downey
- Department of Medicine, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Haley E Randolph
- Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, IL, USA
| | - Erwan Pernet
- Department of Medicine, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Kim A Tran
- Department of Medicine, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Shabaana A Khader
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Irah L King
- Department of Medicine, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Luis B Barreiro
- Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, IL, USA; Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Maziar Divangahi
- Department of Medicine, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada.
| |
Collapse
|
37
|
Nishimura N, Yokota M, Kurihara S, Iwata A, Kageyama T, Ito T, Saku A, Maezawa Y, Hirose K, Nakajima H. Airway epithelial STAT3 inhibits allergic inflammation via upregulation of stearoyl-CoA desaturase 1. Allergol Int 2022; 71:520-527. [PMID: 35660131 DOI: 10.1016/j.alit.2022.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/19/2022] [Accepted: 04/02/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Airway epithelial cells (AECs) play a crucial role in the induction and development of allergic inflammation through the development and activation of immune cells, including Th2 cells and ILC2s. Recent studies have revealed that STAT3 expressed in epithelial cells protects against pathogens and maintains homeostasis in the intestine. However, the roles of STAT3 in airway epithelium are poorly understood. Therefore, we sought to elucidate the roles of airway epithelial STAT3 in allergic airway inflammation. METHODS Allergic airway inflammation was induced by intratracheal administration of house dust mite (HDM) extract in doxycycline-induced AEC-specific STAT3-deficient (STAT3-cKO) mice and their genetic control (STAT3-WT) mice. Airway inflammation was evaluated by flow cytometric analysis of bronchoalveolar lavage fluid cells and histological analysis of the lung. Purified airway epithelial cells were analyzed by quantitative PCR and RNA-sequencing (RNA-seq). RESULTS HDM-induced airway inflammation was exacerbated in STAT3-cKO mice compared with STAT3-WT mice. RNA-seq analyses revealed that Scd1, coding stearoyl-CoA desaturase 1, was most significantly upregulated in HDM-treated STAT3-WT mice compared to HDM-treated STAT3-cKO mice. Notably, the administration of an SCD1 inhibitor exacerbated HDM-induced airway inflammation. AECs of HDM-treated STAT3-cKO mice and those of HDM-treated SCD1 inhibitor-injected mice shared 45 differentially expressed genes (DEGs). Gene enrichment analysis of the DEGs revealed that the enriched ontology clusters included fatty acid biosynthetic process and regulation of lipid biosynthetic process, suggesting the involvement of the STAT3-SCD1-lipid metabolism axis in suppressing allergic inflammation. CONCLUSIONS STAT3 is crucial for suppressing HDM-induced allergic airway inflammation, possibly inducing SCD1 expression in AECs.
Collapse
|
38
|
Smith AP, Williams EP, Plunkett TR, Selvaraj M, Lane LC, Zalduondo L, Xue Y, Vogel P, Channappanavar R, Jonsson CB, Smith AM. Time-Dependent Increase in Susceptibility and Severity of Secondary Bacterial Infections During SARS-CoV-2. Front Immunol 2022; 13:894534. [PMID: 35634338 PMCID: PMC9134015 DOI: 10.3389/fimmu.2022.894534] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/11/2022] [Indexed: 12/20/2022] Open
Abstract
Secondary bacterial infections can exacerbate SARS-CoV-2 infection, but their prevalence and impact remain poorly understood. Here, we established that a mild to moderate infection with the SARS-CoV-2 USA-WA1/2020 strain increased the risk of pneumococcal (type 2 strain D39) coinfection in a time-dependent, but sex-independent, manner in the transgenic K18-hACE2 mouse model of COVID-19. Bacterial coinfection increased lethality when the bacteria was initiated at 5 or 7 d post-virus infection (pvi) but not at 3 d pvi. Bacterial outgrowth was accompanied by neutrophilia in the groups coinfected at 7 d pvi and reductions in B cells, T cells, IL-6, IL-15, IL-18, and LIF were present in groups coinfected at 5 d pvi. However, viral burden, lung pathology, cytokines, chemokines, and immune cell activation were largely unchanged after bacterial coinfection. Examining surviving animals more than a week after infection resolution suggested that immune cell activation remained high and was exacerbated in the lungs of coinfected animals compared with SARS-CoV-2 infection alone. These data suggest that SARS-CoV-2 increases susceptibility and pathogenicity to bacterial coinfection, and further studies are needed to understand and combat disease associated with bacterial pneumonia in COVID-19 patients.
Collapse
Affiliation(s)
- Amanda P. Smith
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Evan P. Williams
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Taylor R. Plunkett
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Muneeswaran Selvaraj
- Department of Acute and Tertiary Care, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Lindey C. Lane
- College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Lillian Zalduondo
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Yi Xue
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Peter Vogel
- Animal Resources Center and Veterinary Pathology Core, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Rudragouda Channappanavar
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Acute and Tertiary Care, University of Tennessee Health Science Center, Memphis, TN, United States
- Institute for the Study of Host-Pathogen Systems, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Colleen B. Jonsson
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
- Institute for the Study of Host-Pathogen Systems, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Amber M. Smith
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
- Institute for the Study of Host-Pathogen Systems, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
39
|
Albayrak N, Orte Cano C, Karimi S, Dogahe D, Van Praet A, Godefroid A, Del Marmol V, Grimaldi D, Bondue B, Van Vooren JP, Mascart F, Corbière V. Distinct Expression Patterns of Interleukin-22 Receptor 1 on Blood Hematopoietic Cells in SARS-CoV-2 Infection. Front Immunol 2022; 13:769839. [PMID: 35422799 PMCID: PMC9004465 DOI: 10.3389/fimmu.2022.769839] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
The new pandemic virus SARS-CoV-2 is characterized by uncontrolled hyper-inflammation in severe cases. As the IL-22/IL-22R1 axis was reported to be involved in inflammation during viral infections, we characterized the expression of IL-22 receptor1, IL-22 and IL-22 binding protein in COVID-19 patients. Blood samples were collected from 19 non-severe and 14 severe patients on the day they presented (D0), at D14, and six months later, and from 6 non-infected controls. The IL-22R1 expression was characterized by flow cytometry. Results were related to HLA-DR expression of myeloid cells, to plasma concentrations of different cytokines and chemokines and NK cells and T lymphocytes functions characterized by their IFN-γ, IL-22, IL-17A, granzyme B and perforin content. The numbers of IL-22R1+ classical, intermediate, and non-classical monocytes and the proportions of IL-22R1+ plasmacytoid DC (pDC), myeloid DC1 and DC2 (mDC1, mDC2) were higher in patients than controls at D0. The proportions of IL-22R1+ classical and intermediate monocytes, and pDC and mDC2 remained high for six months. High proportions of IL-22R1+ non-classical monocytes and mDC2 displayed HLA-DRhigh expression and were thus activated. Multivariate analysis for all IL-22R1+ myeloid cells discriminated the severity of the disease (AUC=0.9023). However, correlation analysis between IL-22R1+ cell subsets and plasma chemokine concentrations suggested pro-inflammatory effects of some subsets and protective effects of others. The numbers of IL-22R1+ classical monocytes and pDC were positively correlated with pro-inflammatory chemokines MCP-1 and IP-10 in severe infections, whereas IL-22R1+ intermediate monocytes were negatively correlated with IL-6, IFN-α and CRP in non-severe infections. Moreover, in the absence of in vitro stimulation, NK and CD4+ T cells produced IFN-γ and IL-22, and CD4+ and CD8+ T cells produced IL-17A. CD4+ T lymphocytes also expressed IL-22R1, the density of its expression defining two different functional subsets. In conclusion, we provide the first evidence that SARS-CoV-2 infection is characterized by an abnormal expression of IL22R1 on blood myeloid cells and CD4+ T lymphocytes. Our results suggest that the involvement of the IL-22R1/IL-22 axis could be protective at the beginning of SARS-CoV-2 infection but could shift to a detrimental response over time.
Collapse
Affiliation(s)
- Nurhan Albayrak
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles, Brussels, Belgium
| | - Carmen Orte Cano
- Department of Dermatology, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Sina Karimi
- Department of Internal Medicine, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - David Dogahe
- Department of Internal Medicine, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Anne Van Praet
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles, Brussels, Belgium
| | - Audrey Godefroid
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles, Brussels, Belgium
| | - Véronique Del Marmol
- Department of Dermatology, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - David Grimaldi
- Department of Intensive Care Unit, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Benjamin Bondue
- Department of Pneumology, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Paul Van Vooren
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles, Brussels, Belgium.,Immunodeficiency Unit, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Françoise Mascart
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles, Brussels, Belgium
| | - Véronique Corbière
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
40
|
Cagan E, Tezcan G, Simsek A, Kizmaz MA, Dombaz F, Asan A, Demir HI, Bal H, Yoyen Ermis D, Gorek Dilektasli A, Kazak E, Akalin EH, Oral HB, Budak F. The Age-Dependent Role of Th22, Tc22, and Tc17 Cells in the Severity of Pneumonia in COVID-19 Immunopathogenesis. Viral Immunol 2022; 35:318-327. [PMID: 35363081 DOI: 10.1089/vim.2021.0132] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) has clinical manifestations ranging from mild symptoms to respiratory failure, septic shock, and multi-organ failure. Lymphocytes are divided into different subtypes based on their cytokine production pattern. In this study, we investigated the role of cytokine expressions of CD4+ T (T helper [Th]1, Th2, Th17, Th22) and CD8+ T cell subtypes (T cytotoxic [Tc]1, Tc2, Tc17, Tc22) in the pathogenesis of COVID-19. Peripheral blood mononuclear cells (PBMCs) were extracted with Ficoll by density gradient centrifugation from blood samples of 180 COVID-19 patients (children and adults) and 30 healthy controls. PBMCs were stimulated with PMA and Ionomycin and treated with Brefeldin A in the fourth hour, and a 10-colored monoclonal antibody panel was evaluated at the end of the sixth hour using flow cytometry. According to our findings, the numbers of Th22 (CD3+, CD4+, and interleukin [IL]-22+) and Tc22 (CD3+, CD8+, IL-22+) cells increased in adult patients regardless of the level of pneumonia (mild, severe, or symptom-free) as compared with healthy controls (p < 0.05). In addition, the number of Tc17 (CD3+, CD8+, and IL-17A+) cells increased in low pneumonia and severe pneumonia groups compared with the healthy controls (p < 0.05). Both IL-22 and IL-17A production decreased during a follow-up within 6 weeks of discharge. Our findings suggest that the increase in only IL-22 expressed Tc22 cells in the 0-12 age group with a general symptom-free course and higher levels of Th22 and Tc22 in uncomplicated adult cases may indicate the protective effect of IL-22. On the contrary, the association between the severity of pneumonia and the elevation of Tc17 cells in adults may reveal the damaging effect of IL-22 when it is co-expressed with IL-17.
Collapse
Affiliation(s)
- Eren Cagan
- Department of Immunology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey.,Department of Pediatric Infectious Diseases, Bursa Yüksek Ihtisas Training and Research Hospital, Health Sciences University, Bursa, Turkey
| | - Gulcin Tezcan
- Department of Fundamental Sciences, Faculty of Dentistry, Bursa Uludag University, Bursa, Turkey
| | - Abdurrahman Simsek
- Department of Immunology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey.,Institution of Health Sciences, Department of Immunology, Bursa Uludag University, Bursa, Turkey
| | - Muhammed Ali Kizmaz
- Department of Immunology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey.,Institution of Health Sciences, Department of Immunology, Bursa Uludag University, Bursa, Turkey
| | - Fatma Dombaz
- Department of Immunology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey.,Institution of Health Sciences, Department of Immunology, Bursa Uludag University, Bursa, Turkey
| | - Ali Asan
- Department of Infectious Diseases, Bursa Yuksek Ihtisas Training and Research Hospital, Health Sciences University, Bursa, Turkey
| | - H Ibrahim Demir
- Department of Immunology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey.,Institution of Health Sciences, Department of Immunology, Bursa Uludag University, Bursa, Turkey
| | - Haldun Bal
- Department of Immunology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Digdem Yoyen Ermis
- Department of Immunology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Aslı Gorek Dilektasli
- Department of Pulmonary Medicine, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Esra Kazak
- Department of Clinical Microbiology and Infection Diseases, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - E Halis Akalin
- Department of Clinical Microbiology and Infection Diseases, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - H Barbaros Oral
- Department of Immunology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Ferah Budak
- Department of Immunology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| |
Collapse
|
41
|
Kadota T, Fujita Y, Araya J, Ochiya T, Kuwano K. Extracellular vesicle-mediated cellular crosstalk in lung repair, remodelling and regeneration. Eur Respir Rev 2022; 31:31/163/210106. [PMID: 35082125 DOI: 10.1183/16000617.0106-2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 10/08/2021] [Indexed: 02/06/2023] Open
Abstract
The unperturbed lung is highly quiescent, with a remarkably low level of cell turnover. However, once damaged, the lung shows an extensive regenerative capacity, with resident progenitor cell populations re-entering the cell cycle and differentiating to promote repair. This quick and dramatic repair response requires interactions among more than 40 different cell lineages in the lung, and defects in any of these processes can lead to various lung pathologies. Understanding the mechanisms of interaction in lung injury, repair and regeneration thus has considerable practical and therapeutic implications. Moreover, therapeutic strategies for replacing lung progenitor cells and their progeny through cell therapy have gained increasing attention. In the last decade, extracellular vesicles (EVs), including exosomes, have been recognised as paracrine mediators through the transfer of biological cargo. Recent work has revealed that EVs are involved in lung homeostasis and diseases. In addition, EVs derived from specific cells or tissues have proven to be a promising cell-free modality for the treatment of lung diseases. This review highlights the EV-mediated cellular crosstalk that regulates lung homeostasis and discusses the potential of EV therapeutics for lung regenerative medicine.
Collapse
Affiliation(s)
- Tsukasa Kadota
- Division of Respiratory Diseases, Dept of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan.,Dept of Translational Research for Exosomes, The Jikei University School of Medicine, Tokyo, Japan
| | - Yu Fujita
- Division of Respiratory Diseases, Dept of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan .,Dept of Translational Research for Exosomes, The Jikei University School of Medicine, Tokyo, Japan
| | - Jun Araya
- Division of Respiratory Diseases, Dept of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Takahiro Ochiya
- Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Kazuyoshi Kuwano
- Division of Respiratory Diseases, Dept of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
42
|
Smith AP, Williams EP, Plunkett TR, Selvaraj M, Lane LC, Zalduondo L, Xue Y, Vogel P, Channappanavar R, Jonsson CB, Smith AM. Time-Dependent Increase in Susceptibility and Severity of Secondary Bacterial Infection during SARS-CoV-2 Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.02.28.482305. [PMID: 35262077 PMCID: PMC8902874 DOI: 10.1101/2022.02.28.482305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Secondary bacterial infections can exacerbate SARS-CoV-2 infection, but their prevalence and impact remain poorly understood. Here, we established that a mild to moderate SARS-CoV-2 infection increased the risk of pneumococcal coinfection in a time-dependent, but sexindependent, manner in the transgenic K18-hACE mouse model of COVID-19. Bacterial coinfection was not established at 3 d post-virus, but increased lethality was observed when the bacteria was initiated at 5 or 7 d post-virus infection (pvi). Bacterial outgrowth was accompanied by neutrophilia in the groups coinfected at 7 d pvi and reductions in B cells, T cells, IL-6, IL-15, IL-18, and LIF were present in groups coinfected at 5 d pvi. However, viral burden, lung pathology, cytokines, chemokines, and immune cell activation were largely unchanged after bacterial coinfection. Examining surviving animals more than a week after infection resolution suggested that immune cell activation remained high and was exacerbated in the lungs of coinfected animals compared with SARS-CoV-2 infection alone. These data suggest that SARS-CoV-2 increases susceptibility and pathogenicity to bacterial coinfection, and further studies are needed to understand and combat disease associated with bacterial pneumonia in COVID-19 patients.
Collapse
Affiliation(s)
- Amanda P. Smith
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Evan P. Williams
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Taylor R. Plunkett
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Muneeswaran Selvaraj
- Department of Acute and Tertiary Care, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Lindey C. Lane
- College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Lillian Zalduondo
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Yi Xue
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Peter Vogel
- Animal Resources Center and Veterinary Pathology Core, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Rudragouda Channappanavar
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Acute and Tertiary Care, University of Tennessee Health Science Center, Memphis, TN, USA
- Institute for the Study of Host-Pathogen Systems, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Colleen B. Jonsson
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
- Institute for the Study of Host-Pathogen Systems, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Amber M. Smith
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
- Institute for the Study of Host-Pathogen Systems, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
43
|
Tissue-resident immunity in the lung: a first-line defense at the environmental interface. Semin Immunopathol 2022; 44:827-854. [PMID: 36305904 PMCID: PMC9614767 DOI: 10.1007/s00281-022-00964-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 09/08/2022] [Indexed: 12/15/2022]
Abstract
The lung is a vital organ that incessantly faces external environmental challenges. Its homeostasis and unimpeded vital function are ensured by the respiratory epithelium working hand in hand with an intricate fine-tuned tissue-resident immune cell network. Lung tissue-resident immune cells span across the innate and adaptive immunity and protect from infectious agents but can also prove to be pathogenic if dysregulated. Here, we review the innate and adaptive immune cell subtypes comprising lung-resident immunity and discuss their ontogeny and role in distinct respiratory diseases. An improved understanding of the role of lung-resident immunity and how its function is dysregulated under pathological conditions can shed light on the pathogenesis of respiratory diseases.
Collapse
|
44
|
Makris S, de Winde CM, Horsnell HL, Cantoral-Rebordinos JA, Finlay RE, Acton SE. Immune function and dysfunction are determined by lymphoid tissue efficacy. Dis Model Mech 2022; 15:dmm049256. [PMID: 35072206 PMCID: PMC8807573 DOI: 10.1242/dmm.049256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Lymphoid tissue returns to a steady state once each immune response is resolved, and although this occurs multiple times throughout life, its structural integrity and functionality remain unaffected. Stromal cells orchestrate cellular interactions within lymphoid tissue, and any changes to the microenvironment can have detrimental outcomes and drive disease. A breakdown in lymphoid tissue homeostasis can lead to a loss of tissue structure and function that can cause aberrant immune responses. This Review highlights recent advances in our understanding of lymphoid tissue function and remodelling in adaptive immunity and in disease states. We discuss the functional role of lymphoid tissue in disease progression and explore the changes to lymphoid tissue structure and function driven by infection, chronic inflammatory conditions and cancer. Understanding the role of lymphoid tissues in immune responses to a wide range of pathologies allows us to take a fuller systemic view of disease progression.
Collapse
Affiliation(s)
- Spyridon Makris
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Charlotte M. de Winde
- Department for Molecular Cell Biology and Immunology, Amsterdam UMC, location VUmc, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands
| | - Harry L. Horsnell
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Jesús A. Cantoral-Rebordinos
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Rachel E. Finlay
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester M13 9PL, UK
| | - Sophie E. Acton
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
45
|
Kumar A, Patel VS, Harding JN, You D, Cormier SA. Exposure to combustion derived particulate matter exacerbates influenza infection in neonatal mice by inhibiting IL22 production. Part Fibre Toxicol 2021; 18:43. [PMID: 34906172 PMCID: PMC8670221 DOI: 10.1186/s12989-021-00438-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 11/29/2021] [Indexed: 01/05/2023] Open
Abstract
Background Particulate matter (PM) containing environmentally persistent free radicals (EPFRs) are formed during various combustion processes, including the thermal remediation of hazardous wastes. Exposure to PM adversely affects respiratory health in infants and is associated with increased morbidity and mortality due to acute lower respiratory tract infections. We previously reported that early-life exposure to PM damages the lung epithelium and suppresses immune responses to influenza virus (Flu) infection, thereby enhancing Flu severity. Interleukin 22 (IL22) is important in resolving lung injury following Flu infection. In the current study, we determined the effects of PM exposure on pulmonary IL22 responses using our neonatal mouse model of Flu infection. Results Exposure to PM resulted in an immediate (0.5–1-day post-exposure; dpe) increase in IL22 expression in the lungs of C57BL/6 neonatal mice; however, this IL22 expression was not maintained and failed to increase with either continued exposure to PM or subsequent Flu infection of PM-exposed mice. This contrasts with increased IL22 expression in age-matched mice exposed to vehicle and Flu infected. Activation of the aryl hydrocarbon receptor (AhR), which mediates the induction and release of IL22 from immune cells, was also transiently increased with PM exposure. The microbiome plays a major role in maintaining epithelial integrity and immune responses by producing various metabolites that act as ligands for AhR. Exposure to PM induced lung microbiota dysbiosis and altered the levels of indole, a microbial metabolite. Treatment with recombinant IL22 or indole-3-carboxaldehyde (I3A) prevented PM associated lung injury. In addition, I3A treatment also protected against increased mortality in Flu-infected mice exposed to PMs. Conclusions Together, these data suggest that exposure to PMs results in failure to sustain IL22 levels and an inability to induce IL22 upon Flu infection. Insufficient levels of IL22 may be responsible for aberrant epithelial repair and immune responses, leading to increased Flu severity in areas of high PM.
Collapse
Affiliation(s)
- Avinash Kumar
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA.,Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, USA
| | - Vivek S Patel
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - Jeffrey N Harding
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - Dahui You
- Department of Pediatrics, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Stephania A Cormier
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA. .,Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, USA.
| |
Collapse
|
46
|
Me R, Gao N, Zhang Y, Lee PSY, Wang J, Liu T, Standiford TJ, Mi QS, Yu FSX. IL-36α Enhances Host Defense against Pseudomonas aeruginosa Keratitis in C57BL/6 Mouse Corneas. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:2868-2877. [PMID: 34686582 PMCID: PMC8612993 DOI: 10.4049/jimmunol.2001246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 09/21/2021] [Indexed: 12/13/2022]
Abstract
The IL-36 cytokines are known to play various roles in mediating the immune response to infection in a tissue- and pathogen-dependent manner. The present study seeks to investigate the role of IL-36R signaling in C57BL/6 mouse corneas in response to Pseudomonas aeruginosa infection. IL-36α-/-, IL-36γ-/-, and IL-36R-/- mice had significantly more severe keratitis than wild-type mice. At six hours postinfection, IL-36α pretreatment augmented P. aeruginosa-induced expression of IL-1Ra, IL-36γ, LCN2, and S100A8/A9. At one day postinfection, exogenous IL-36α suppressed, whereas IL-36α deficiency promoted, the expression of IL-1β. At three days postinfection, exogenous IL-36α suppressed Th1 but promoted Th2 immune response. IL-36α stimulated the infiltration of IL-22-expressing immune cells, and IL-22 neutralization resulted in more severe keratitis. IL-36α alone stimulated dendritic cell infiltration in B6 mouse corneas. Taken together, our study suggests that IL-36R signaling plays a protective role in the pathogenesis of P. aeruginosa keratitis by promoting the innate immune defense, Th2, and/or Th22/IL-22 immune responses. Exogenous IL-36α might be a potential therapy for improving the outcome of P. aeruginosa keratitis.
Collapse
Affiliation(s)
- Rao Me
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI
| | - Nan Gao
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI
| | - Yangyang Zhang
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI
| | - Patrick S Y Lee
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI
| | - Jie Wang
- Center for Cutaneous Biology and Immunology, Department of Dermatology and Immunology Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI; and
| | - Tingting Liu
- Center for Cutaneous Biology and Immunology, Department of Dermatology and Immunology Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI; and
| | - Theodore J Standiford
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI
| | - Qing-Sheng Mi
- Center for Cutaneous Biology and Immunology, Department of Dermatology and Immunology Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI; and
| | - Fu-Shin X Yu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI;
| |
Collapse
|
47
|
Barman TK, Metzger DW. Disease Tolerance during Viral-Bacterial Co-Infections. Viruses 2021; 13:v13122362. [PMID: 34960631 PMCID: PMC8706933 DOI: 10.3390/v13122362] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/29/2021] [Accepted: 11/23/2021] [Indexed: 11/16/2022] Open
Abstract
Disease tolerance has emerged as an alternative way, in addition to host resistance, to survive viral-bacterial co-infections. Disease tolerance plays an important role not in reducing pathogen burden, but in maintaining tissue integrity and controlling organ damage. A common co-infection is the synergy observed between influenza virus and Streptococcus pneumoniae that results in superinfection and lethality. Several host cytokines and cells have shown promise in promoting tissue protection and damage control while others induce severe immunopathology leading to high levels of morbidity and mortality. The focus of this review is to describe the host cytokines and innate immune cells that mediate disease tolerance and lead to a return to host homeostasis and ultimately, survival during viral-bacterial co-infection.
Collapse
|
48
|
Taghavi S, Jackson-Weaver O, Abdullah S, Wanek A, Drury R, Packer J, Cotton-Betteridge A, Duchesne J, Pociask D, Kolls J. Interleukin-22 mitigates acute respiratory distress syndrome (ARDS). PLoS One 2021; 16:e0254985. [PMID: 34597299 PMCID: PMC8486146 DOI: 10.1371/journal.pone.0254985] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 07/07/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The goal of this study was to determine if IL-22:Fc would Acute Respiratory Distress Syndrome (ARDS). SUMMARY BACKGROUND DATA No therapies exist for ARDS and treatment is purely supportive. Interleukin-22 (IL-22) plays an integral component in recovery of the lung from infection. IL-22:Fc is a recombinant protein with a human FC immunoglobulin that increases the half-life of IL-22. STUDY DESIGN ARDS was induced in C57BL/6 mice with intra-tracheal lipopolysaccharide (LPS) at a dose of 33.3 or 100 ug. In the low-dose LPS group (LDG), IL-22:FC was administered via tail vein injection at 30 minutes (n = 9) and compared to sham (n = 9). In the high-dose LPS group (HDG), IL-22:FC was administered (n = 11) then compared to sham (n = 8). Euthanasia occurred after bronchioalveolar lavage (BAL) on post-injury day 4. RESULTS In the LDG, IL-22:FC resulted in decreased protein leak (0.15 vs. 0.25 ug/uL, p = 0.02). BAL protein in animals receiving IL-22:Fc in the HDG was not different. For the HDG, animals receiving IL-22:Fc had lower BAL cell counts (539,636 vs 3,147,556 cells/uL, p = 0.02). For the HDG, IL-6 (110.6 vs. 527.1 pg/mL, p = 0.04), TNF-α (5.87 vs. 25.41 pg/mL, p = 0.04), and G-CSF (95.14 vs. 659.6, p = 0.01) levels were lower in the BAL fluid of IL-22:Fc treated animals compared to sham. CONCLUSIONS IL-22:Fc decreases lung inflammation and lung capillary leak in ARDS. IL-22:Fc may be a novel therapy for ARDS.
Collapse
Affiliation(s)
- Sharven Taghavi
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, United States of America
| | - Olan Jackson-Weaver
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, United States of America
| | - Sarah Abdullah
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, United States of America
| | - Alanna Wanek
- Tulane University School of Medicine, Center for Translational Research in Infection and Inflammation, New Orleans, LA, United States of America
| | - Robert Drury
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, United States of America
| | - Jacob Packer
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, United States of America
| | - Aaron Cotton-Betteridge
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, United States of America
| | - Juan Duchesne
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, United States of America
| | - Derek Pociask
- Tulane University School of Medicine, Center for Translational Research in Infection and Inflammation, New Orleans, LA, United States of America
| | - Jay Kolls
- Tulane University School of Medicine, Center for Translational Research in Infection and Inflammation, New Orleans, LA, United States of America
| |
Collapse
|
49
|
Toward a universal influenza virus vaccine: Some cytokines may fulfill the request. Cytokine 2021; 148:155703. [PMID: 34555604 DOI: 10.1016/j.cyto.2021.155703] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 01/21/2023]
Abstract
The influenza virus annually causes widespread damages to the health and economy of the global community. Vaccination is currently the most crucial strategy in reducing the number of patients. Genetic variations, the high diversity of pandemic viruses, and zoonoses make it challenging to select suitable strains for annual vaccine production. If new pandemic viruses emerge, it will take a long time to produce a vaccine according to the new strains. In the present study, intending to develop a universal influenza vaccine, new bicistronic DNA vaccines were developed that expressed NP or NPm antigen with one of modified IL-18/ IL-17A/ IL-22 cytokine adjuvants. NPm is a mutant form of the antigen that has the ability for cytoplasmic accumulation. In order to investigate and differentiate the role of each of the components of Th1, Th2, Th17, and Treg cellular immune systems in the performance of vaccines, Treg competent and Treg suppressed mouse groups were used. Mice were vaccinated with Foxp3-FC immunogen to produce Treg suppressed mouse groups. The potential of the vaccines to stimulate the immune system was assessed by IFN-γ/IL-17A Dual FluoroSpot. The vaccine's ability to induce humoral immune response was determined by measuring IgG1, IgG2a, and IgA-specific antibodies against the antigen. Kinetics of Th1, Th2, and Th17 cellular immune responses after vaccination, were assessed by evaluating the expression changes of IL-17A, IFN-γ, IL-18, IL-22, IL-4, and IL-2 cytokines by semi-quantitative real-time RT-PCR. To assess the vaccines' ability to induce heterosubtypic immunity, challenge tests with homologous and heterologous viruses were performed and then the virus titer was measured in the lungs of animals. Evaluation of the data obtained from this study showed that the DNA-vaccines coding NPm have more ability to induces a potent cross-cellular immune response and protective immunity than DNA-vaccines coding NP. Although the use of IL-18/ IL-17A/ IL-22 genetic adjuvants enhanced immune responses and protective immunity, Administration of NPm in combination with modified IL-18 (Igk-mIL18-IgFC) induced the most effective immunity in Treg competent mice group.
Collapse
|
50
|
Viral and Bacterial Co-Infections in the Lungs: Dangerous Liaisons. Viruses 2021; 13:v13091725. [PMID: 34578306 PMCID: PMC8472850 DOI: 10.3390/v13091725] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/23/2022] Open
Abstract
Respiratory tract infections constitute a significant public health problem, with a therapeutic arsenal that remains relatively limited and that is threatened by the emergence of antiviral and/or antibiotic resistance. Viral–bacterial co-infections are very often associated with the severity of these respiratory infections and have been explored mainly in the context of bacterial superinfections following primary influenza infection. This review summarizes our current knowledge of the mechanisms underlying these co-infections between respiratory viruses (influenza viruses, RSV, and SARS-CoV-2) and bacteria, at both the physiological and immunological levels. This review also explores the importance of the microbiome and the pathological context in the evolution of these respiratory tract co-infections and presents the different in vitro and in vivo experimental models available. A better understanding of the complex functional interactions between viruses/bacteria and host cells will allow the development of new, specific, and more effective diagnostic and therapeutic approaches.
Collapse
|